Signaling by PTK6 (Homo sapiens)

From WikiPathways

Jump to: navigation, search
8, 11, 21, 22, 24...7643226322, 1074021, 72148, 166436021, 727411574, 86, 163162246311514216246, 63, 10853136, 16323, 109, 151, 160727491, 148, 16653761076378634010932, 53, 1341421071154377, 1101104336, 13626, 78434311512011, 12032cytosolnucleoplasmEGFR CCNE1 ADPGDP p-Y342-PTK6 STAT3PTK6 ADPDOK1 UBC(533-608) CCNE1 S-Farn-Me-PalmS KRAS4A p-Y342-PTK6 RHOA S-Farn-Me KRAS4B S-Farn-Me PalmS NRAS CDK4 p-Y705-STAT3dimer:SOCS3 GenePTK6 Genep-Y-KHDRBS2p-Y705-STAT3RAC1:GTPSOCS3RHO GTPases activateCITKHDRSB3S-Farn-Me-2xPalmS HRAS LRRK2SRMSADPCCND1 UBC(381-456) ATPBCAR1 RASA1 SemaphorininteractionsRHO GTPases activatePKNsp-Y525-GPNMB STAP2 p-Y342-PTK6 STAP2DOK1ADPPip-Y1105-ARHGAP35 CCNE1 ATPp-Y342-PTK6 p-Y351-PTK6 ATPRHO GTPases activateKTN1ATPADPp-Y88-CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))GTP p-Y88-CDKN1B p-Y342-PTK6:ARHGAP35p21 RAS:GTPADPp-Y31,Y118-PXN:CRK:DOCK180:ELMO1,ELMO2NR3C1 ADPCRK p-Y342-PTK6:DOK1EPAS1 EGFR p-Y342-PTK6:STAP2UBC(1-76) ARAP1 HBEGF(63-148) p-Y705-STAT3 CDKN1B p-Y342-PTK6 ELMO1 PELP1 UBC(153-228) p-Y435,Y440,Y443-KHDRBS1NR3C1:(ALDO,11DCORST,CORST,CORT) dimerGTP HBEGF(63-148) GTP ADPRHO GTPases activatePAKsARHGAP35 RAC1:GDPUBB(1-76) GDPEGFR KHDRBS1PolyUb,p-Y700,Y731,Y774-CBLHIF1A CRK:DOCK180:ELMO1,ELMO2SOCS3 p-Y31,Y118-PXNS-Farn-Me KRAS4B ALDO p-Y1105-ARHGAP35UBC(609-684) p-Y705-STAT3 dimerp-Y231-ARAP1p-Y342-PTK6:AKT1p-ERBB2 heterodimersPip-Y342-PTK6 p-Y342-PTK6:CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))p-Y525-GPNMB CDK4 DOCK1 CCND1 RAF/MAP kinasecascadep-Y1105-ARHGAP35:RASA1S-Farn-Me-PalmS KRAS4A ARAP1GDP CDK2 Mitotic G1 phase andG1/S transitionATPATPRAC1 CDK4 ATPLINC01139 S-Farn-Me-2xPalmS HRAS PTPN1CORT ALDO AKT1RHO GTPases ActivateWASPs and WAVEsp-Y525-GPNMB PIP3 activates AKTsignalingp-Y342,Y447-PTK6CCND1 p21 RAS:GTP:RASA1p-Y342-PTK6ATPPTK6 Signaling by ERBB2p-Y342-PTK6:p-Y315,Y326-AKT1Signaling by EGFRRASA1 RPS27A(1-76) p-Y700,Y731,Y774-CBLp-Y-KHDRSB3ADPRHO GTPases ActivateForminsATPRHOA:GTPp-Y565,S797-HIF1AS PhaseRHOA p-Y362-DOK1EPAS1 GTPp-Y342-PTK6:ARAP1RAC1 11DCORST SOCS3 Genep-Y342-PTK6 HBEGF:EGFRS-Farn-Me KRAS4B UBC(229-304) PTK6EGFR UBC(77-152) EGFR UBC(457-532) HIF1A STAT3 ADPp-Y250-STAP2 RASA1ATPBCAR1HBEGF(63-148) p-Y342-PTK6 ATPp-Y250-STAP2 p-Y342-PTK6 UBC(305-380) HBEGF:EGFR:p-Y525-GPNMB:LINC01139:PTK6:LRRK2Ubp-6Y-ERBB2 heterodimers S-Farn-Me-PalmS KRAS4A UBA52(1-76) p-Y565-HIF1AELMO2 GTP PTPN1 p-Y342-PTK6:p-Y250-STAP2:STAT3PTK6 Gene ARHGAP35ELMO2 Autophosphorylated p-Y877-ERBB2 heterodimers ATPRHOA S-Farn-Me PalmS NRAS p-Y342-PTK6:PXNp-Y342-PTK6 ADPp-Y342-PTK6:p-Y250-STAP2PXN ATPHBEGF:EGFR:p-Y525-GPNMB:LINC01139:p-Y351-PTK6:LRRK2RHO GTPases activateIQGAPsCRK ADPPXNHBEGF:EGFR:GPNMBADPPTK6Gene:EPAS1:NR3C1:Glucocorticoid ligand:PELP1ATPp-Y315,Y326-AKT1 ATPKHDRBS2S-Farn-Me PalmS NRAS NR3C1 LRRK2 H2Op-Y342-PTK6 PELP1HBEGF(63-148) CDK2 EPAS1 CBLHIF1A,EPAS1:PTK6Genep21 RAS:GDPRHOA:GDPGDP p-Y-SFPQp-Y435,Y440,Y443-KHDRBS1p-Y342-PTK6:PTPN1p-Y342-PTK6p-Y1105-ARHGAP35 ELMO1 p-Y705-STAT3 Autophosphorylated p-Y877-ERBB2 heterodimers CORST GPNMBDOCK1 SOCS3 Gene HBEGF(63-148) AKT1 ADPp-Y342-PTK6:SOCS3SFPQCDK2 CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))HBEGF:EGFR:p-Y525-GPNMBCellular response tohypoxiaCDKN1B p-Y705-STAT3 dimerCORST HIF1A,EPAS1p-Y342-PTK6:BCAR1LINC01139p-Y342-PTK6 GTP p-Y342-PTK6 p-Y705-STAT3 LRRK2 p-6Y-ERBB2 heterodimers p-ERBB2heterodimers:PTK6p-Y165,Y664-BCAR1UBB(153-228) p-Y31,Y118-PXN p-Y342-PTK6:KHDRBS1p-Y1105-ARHGAP35:RHOA:GTPCORT HIF1ALINC01139 PTK6 Gene ATPADPS-Farn-Me-2xPalmS HRAS UBB(77-152) p-Y342-PTK6 ADP11DCORST KHDRBS1 EPAS1GPNMB 26, 787819, 38531667, 10, 18, 106, 119...1071401623, 4, 62, 71, 75...2, 13, 34, 45, 51...6, 20, 30, 35, 52...4312, 17, 29, 49, 58...14, 15, 31, 33, 42...9, 54, 55, 61, 66...115431, 25, 146, 164431152274, 862136405, 16, 28, 50, 1611107443766346, 632257, 79, 83, 85, 97...6372


Description

PTK6 (BRK) is an oncogenic non-receptor tyrosine kinase that functions downstream of ERBB2 (HER2) (Xiang et al. 2008, Peng et al. 2015) and other receptor tyrosine kinases, such as EGFR (Kamalati et al. 1996) and MET (Castro and Lange 2010). Since ERBB2 forms heterodimers with EGFR and since MET can heterodimerize with both ERBB2 and EGFR (Tanizaki et al. 2011), it is not clear if MET and EGFR activate PTK6 directly or act through ERBB2. Levels of PTK6 increase under hypoxic conditions (Regan Anderson et al. 2013, Pires et al. 2014). The kinase activity of PTK6 is negatively regulated by PTPN1 phosphatase (Fan et al. 2013) and SRMS kinase (Fan et al. 2015), as well as the STAT3 target SOCS3 (Gao et al. 2012).

PTK6 activates STAT3-mediated transcription (Ikeda et al. 2009, Ikeda et al. 2010) and may also activate STAT5-mediated transcription (Ikeda et al. 2011). PTK6 promotes cell motility and migration by regulating the activity of RHO GTPases RAC1 (Chen et al. 2004) and RHOA (Shen et al. 2008), and possibly by affecting motility-related kinesins (Lukong and Richard 2008). PTK6 crosstalks with AKT1 (Zhang et al. 2005, Zheng et al. 2010) and RAS signaling cascades (Shen et al. 2008, Ono et al. 2014) and may be involved in MAPK7 (ERK5) activation (Ostrander et al. 2007, Zheng et al. 2012). PTK6 enhances EGFR signaling by inhibiting EGFR down-regulation (Kang et al. 2010, Li et al. 2012, Kang and Lee 2013). PTK6 may also enhance signaling by IGF1R (Fan et al. 2013) and ERBB3 (Kamalati et al. 2000).<p>PTK6 promotes cell cycle progression by phosphorylating and inactivating CDK inhibitor CDKN1B (p27) (Patel et al. 2015).<p>PTK6 activity is upregulated in osteopontin (OPN or SPP1)-mediated signaling, leading to increased VEGF expression via PTK6/NF-kappaB/ATF4 signaling path. PTK6 may therefore play a role in VEGF-dependent tumor angiogenesis (Chakraborty et al. 2008).<p>PTK6 binds and phosphorylates several nuclear RNA-binding proteins, including SAM68 family members (KHDRSB1, KHDRSB2 and KHDRSB3) (Derry et al. 2000, Haegebarth et al. 2004, Lukong et al. 2005) and SFPQ (PSF) (Lukong et al. 2009). The biological role of PTK6 in RNA processing is not known.<p>For a review of PTK6 function, please refer to Goel and Lukong 2015. View original pathway at Reactome.</div>

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 8848021
Reactome-version 
Reactome version: 75
Reactome Author 
Reactome Author: Orlic-Milacic, Marija

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Carpenter G.; ''Employment of the epidermal growth factor receptor in growth factor-independent signaling pathways.''; PubMed Europe PMC Scholia
  2. Yasuda S, Oceguera-Yanez F, Kato T, Okamoto M, Yonemura S, Terada Y, Ishizaki T, Narumiya S.; ''Cdc42 and mDia3 regulate microtubule attachment to kinetochores.''; PubMed Europe PMC Scholia
  3. Kuroda S, Fukata M, Kobayashi K, Nakafuku M, Nomura N, Iwamatsu A, Kaibuchi K.; ''Identification of IQGAP as a putative target for the small GTPases, Cdc42 and Rac1.''; PubMed Europe PMC Scholia
  4. Pelikan-Conchaudron A, Le Clainche C, Didry D, Carlier MF.; ''The IQGAP1 protein is a calmodulin-regulated barbed end capper of actin filaments: possible implications in its function in cell migration.''; PubMed Europe PMC Scholia
  5. Zhou Y, Gunput RA, Pasterkamp RJ.; ''Semaphorin signaling: progress made and promises ahead.''; PubMed Europe PMC Scholia
  6. Roskoski R.; ''RAF protein-serine/threonine kinases: structure and regulation.''; PubMed Europe PMC Scholia
  7. Majmundar AJ, Wong WJ, Simon MC.; ''Hypoxia-inducible factors and the response to hypoxic stress.''; PubMed Europe PMC Scholia
  8. Kamalati T, Jolin HE, Fry MJ, Crompton MR.; ''Expression of the BRK tyrosine kinase in mammary epithelial cells enhances the coupling of EGF signalling to PI 3-kinase and Akt, via erbB3 phosphorylation.''; PubMed Europe PMC Scholia
  9. Watanabe S, De Zan T, Ishizaki T, Narumiya S.; ''Citron kinase mediates transition from constriction to abscission through its coiled-coil domain.''; PubMed Europe PMC Scholia
  10. Pouysségur J, Dayan F, Mazure NM.; ''Hypoxia signalling in cancer and approaches to enforce tumour regression.''; PubMed Europe PMC Scholia
  11. Pires IM, Blokland NJ, Broos AW, Poujade FA, Senra JM, Eccles SA, Span PN, Harvey AJ, Hammond EM.; ''HIF-1α-independent hypoxia-induced rapid PTK6 stabilization is associated with increased motility and invasion.''; PubMed Europe PMC Scholia
  12. Chittenden T, Livingston DM, Kaelin WG.; ''The T/E1A-binding domain of the retinoblastoma product can interact selectively with a sequence-specific DNA-binding protein.''; PubMed Europe PMC Scholia
  13. Seth A, Otomo C, Rosen MK.; ''Autoinhibition regulates cellular localization and actin assembly activity of the diaphanous-related formins FRLalpha and mDia1.''; PubMed Europe PMC Scholia
  14. Hutchinson CL, Lowe PN, McLaughlin SH, Mott HR, Owen D.; ''Mutational analysis reveals a single binding interface between RhoA and its effector, PRK1.''; PubMed Europe PMC Scholia
  15. Palmer RH, Dekker LV, Woscholski R, Le Good JA, Gigg R, Parker PJ.; ''Activation of PRK1 by phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-trisphosphate. A comparison with protein kinase C isotypes.''; PubMed Europe PMC Scholia
  16. Koncina E, Roth L, Gonthier B, Bagnard D.; ''Role of semaphorins during axon growth and guidance.''; PubMed Europe PMC Scholia
  17. Lees JA, Saito M, Vidal M, Valentine M, Look T, Harlow E, Dyson N, Helin K.; ''The retinoblastoma protein binds to a family of E2F transcription factors.''; PubMed Europe PMC Scholia
  18. Loenarz C, Schofield CJ.; ''Physiological and biochemical aspects of hydroxylations and demethylations catalyzed by human 2-oxoglutarate oxygenases.''; PubMed Europe PMC Scholia
  19. Vignal E, Blangy A, Martin M, Gauthier-Rouvière C, Fort P.; ''Kinectin is a key effector of RhoG microtubule-dependent cellular activity.''; PubMed Europe PMC Scholia
  20. Cantwell-Dorris ER, O'Leary JJ, Sheils OM.; ''BRAFV600E: implications for carcinogenesis and molecular therapy.''; PubMed Europe PMC Scholia
  21. Regan Anderson TM, Peacock DL, Daniel AR, Hubbard GK, Lofgren KA, Girard BJ, Schörg A, Hoogewijs D, Wenger RH, Seagroves TN, Lange CA.; ''Breast tumor kinase (Brk/PTK6) is a mediator of hypoxia-associated breast cancer progression.''; PubMed Europe PMC Scholia
  22. Patel P, Asbach B, Shteyn E, Gomez C, Coltoff A, Bhuyan S, Tyner AL, Wagner R, Blain SW.; ''Brk/Protein tyrosine kinase 6 phosphorylates p27KIP1, regulating the activity of cyclin D-cyclin-dependent kinase 4.''; PubMed Europe PMC Scholia
  23. Ahmadian MR, Stege P, Scheffzek K, Wittinghofer A.; ''Confirmation of the arginine-finger hypothesis for the GAP-stimulated GTP-hydrolysis reaction of Ras.''; PubMed Europe PMC Scholia
  24. Fan G, Aleem S, Yang M, Miller WT, Tonks NK.; ''Protein-tyrosine Phosphatase and Kinase Specificity in Regulation of SRC and Breast Tumor Kinase.''; PubMed Europe PMC Scholia
  25. Herbst RS.; ''Review of epidermal growth factor receptor biology.''; PubMed Europe PMC Scholia
  26. Zhang P, Ostrander JH, Faivre EJ, Olsen A, Fitzsimmons D, Lange CA.; ''Regulated association of protein kinase B/Akt with breast tumor kinase.''; PubMed Europe PMC Scholia
  27. Kamalati T, Jolin HE, Mitchell PJ, Barker KT, Jackson LE, Dean CJ, Page MJ, Gusterson BA, Crompton MR.; ''Brk, a breast tumor-derived non-receptor protein-tyrosine kinase, sensitizes mammary epithelial cells to epidermal growth factor.''; PubMed Europe PMC Scholia
  28. Dickson BJ.; ''Molecular mechanisms of axon guidance.''; PubMed Europe PMC Scholia
  29. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR.; ''The E2F transcription factor is a cellular target for the RB protein.''; PubMed Europe PMC Scholia
  30. Cargnello M, Roux PP.; ''Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases.''; PubMed Europe PMC Scholia
  31. Yoshinaga C, Mukai H, Toshimori M, Miyamoto M, Ono Y.; ''Mutational analysis of the regulatory mechanism of PKN: the regulatory region of PKN contains an arachidonic acid-sensitive autoinhibitory domain.''; PubMed Europe PMC Scholia
  32. Lukong KE, Larocque D, Tyner AL, Richard S.; ''Tyrosine phosphorylation of sam68 by breast tumor kinase regulates intranuclear localization and cell cycle progression.''; PubMed Europe PMC Scholia
  33. Maesaki R, Ihara K, Shimizu T, Kuroda S, Kaibuchi K, Hakoshima T.; ''The structural basis of Rho effector recognition revealed by the crystal structure of human RhoA complexed with the effector domain of PKN/PRK1.''; PubMed Europe PMC Scholia
  34. Romero S, Le Clainche C, Didry D, Egile C, Pantaloni D, Carlier MF.; ''Formin is a processive motor that requires profilin to accelerate actin assembly and associated ATP hydrolysis.''; PubMed Europe PMC Scholia
  35. Kyriakis JM, Avruch J.; ''Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update.''; PubMed Europe PMC Scholia
  36. Gao Y, Cimica V, Reich NC.; ''Suppressor of cytokine signaling 3 inhibits breast tumor kinase activation of STAT3.''; PubMed Europe PMC Scholia
  37. Li X, Lu Y, Liang K, Hsu JM, Albarracin C, Mills GB, Hung MC, Fan Z.; ''Brk/PTK6 sustains activated EGFR signaling through inhibiting EGFR degradation and transactivating EGFR.''; PubMed Europe PMC Scholia
  38. Hotta K, Tanaka K, Mino A, Kohno H, Takai Y.; ''Interaction of the Rho family small G proteins with kinectin, an anchoring protein of kinesin motor.''; PubMed Europe PMC Scholia
  39. Castro NE, Lange CA.; ''Breast tumor kinase and extracellular signal-regulated kinase 5 mediate Met receptor signaling to cell migration in breast cancer cells.''; PubMed Europe PMC Scholia
  40. Kang SA, Lee ES, Yoon HY, Randazzo PA, Lee ST.; ''PTK6 inhibits down-regulation of EGF receptor through phosphorylation of ARAP1.''; PubMed Europe PMC Scholia
  41. Goel RK, Lukong KE.; ''Tracing the footprints of the breast cancer oncogene BRK - Past till present.''; PubMed Europe PMC Scholia
  42. Owen D, Lowe PN, Nietlispach D, Brosnan CE, Chirgadze DY, Parker PJ, Blundell TL, Mott HR.; ''Molecular dissection of the interaction between the small G proteins Rac1 and RhoA and protein kinase C-related kinase 1 (PRK1).''; PubMed Europe PMC Scholia
  43. Lin A, Li C, Xing Z, Hu Q, Liang K, Han L, Wang C, Hawke DH, Wang S, Zhang Y, Wei Y, Ma G, Park PK, Zhou J, Zhou Y, Hu Z, Zhou Y, Marks JR, Liang H, Hung MC, Lin C, Yang L.; ''The LINK-A lncRNA activates normoxic HIF1α signalling in triple-negative breast cancer.''; PubMed Europe PMC Scholia
  44. Hutchinson CL, Lowe PN, McLaughlin SH, Mott HR, Owen D.; ''Differential binding of RhoA, RhoB, and RhoC to protein kinase C-related kinase (PRK) isoforms PRK1, PRK2, and PRK3: PRKs have the highest affinity for RhoB.''; PubMed Europe PMC Scholia
  45. Habas R, Kato Y, He X.; ''Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daam1.''; PubMed Europe PMC Scholia
  46. Li R, Zhang B, Zheng Y.; ''Structural determinants required for the interaction between Rho GTPase and the GTPase-activating domain of p190.''; PubMed Europe PMC Scholia
  47. Ostrander JH, Daniel AR, Lofgren K, Kleer CG, Lange CA.; ''Breast tumor kinase (protein tyrosine kinase 6) regulates heregulin-induced activation of ERK5 and p38 MAP kinases in breast cancer cells.''; PubMed Europe PMC Scholia
  48. Dettori R, Sonzogni S, Meyer L, Lopez-Garcia LA, Morrice NA, Zeuzem S, Engel M, Piiper A, Neimanis S, Frödin M, Biondi RM.; ''Regulation of the interaction between protein kinase C-related protein kinase 2 (PRK2) and its upstream kinase, 3-phosphoinositide-dependent protein kinase 1 (PDK1).''; PubMed Europe PMC Scholia
  49. Depoortere F, Van Keymeulen A, Lukas J, Costagliola S, Bartkova J, Dumont JE, Bartek J, Roger PP, Dremier S.; ''A requirement for cyclin D3-cyclin-dependent kinase (cdk)-4 assembly in the cyclic adenosine monophosphate-dependent proliferation of thyrocytes.''; PubMed Europe PMC Scholia
  50. Pasterkamp RJ, Verhaagen J.; ''Semaphorins in axon regeneration: developmental guidance molecules gone wrong?''; PubMed Europe PMC Scholia
  51. Xu Y, Moseley JB, Sagot I, Poy F, Pellman D, Goode BL, Eck MJ.; ''Crystal structures of a Formin Homology-2 domain reveal a tethered dimer architecture.''; PubMed Europe PMC Scholia
  52. Roskoski R.; ''ERK1/2 MAP kinases: structure, function, and regulation.''; PubMed Europe PMC Scholia
  53. Derry JJ, Richard S, Valderrama Carvajal H, Ye X, Vasioukhin V, Cochrane AW, Chen T, Tyner AL.; ''Sik (BRK) phosphorylates Sam68 in the nucleus and negatively regulates its RNA binding ability.''; PubMed Europe PMC Scholia
  54. Gruneberg U, Neef R, Li X, Chan EH, Chalamalasetty RB, Nigg EA, Barr FA.; ''KIF14 and citron kinase act together to promote efficient cytokinesis.''; PubMed Europe PMC Scholia
  55. Serres MP, Kossatz U, Chi Y, Roberts JM, Malek NP, Besson A.; ''p27(Kip1) controls cytokinesis via the regulation of citron kinase activation.''; PubMed Europe PMC Scholia
  56. Mukai H, Toshimori M, Shibata H, Takanaga H, Kitagawa M, Miyahara M, Shimakawa M, Ono Y.; ''Interaction of PKN with alpha-actinin.''; PubMed Europe PMC Scholia
  57. Chan PM, Manser E.; ''PAKs in human disease.''; PubMed Europe PMC Scholia
  58. Cobrinik D.; ''Pocket proteins and cell cycle control.''; PubMed Europe PMC Scholia
  59. Flynn P, Mellor H, Casamassima A, Parker PJ.; ''Rho GTPase control of protein kinase C-related protein kinase activation by 3-phosphoinositide-dependent protein kinase.''; PubMed Europe PMC Scholia
  60. Lukong KE, Huot ME, Richard S.; ''BRK phosphorylates PSF promoting its cytoplasmic localization and cell cycle arrest.''; PubMed Europe PMC Scholia
  61. Madaule P, Furuyashiki T, Reid T, Ishizaki T, Watanabe G, Morii N, Narumiya S.; ''A novel partner for the GTP-bound forms of rho and rac.''; PubMed Europe PMC Scholia
  62. White CD, Erdemir HH, Sacks DB.; ''IQGAP1 and its binding proteins control diverse biological functions.''; PubMed Europe PMC Scholia
  63. Shen CH, Chen HY, Lin MS, Li FY, Chang CC, Kuo ML, Settleman J, Chen RH.; ''Breast tumor kinase phosphorylates p190RhoGAP to regulate rho and ras and promote breast carcinoma growth, migration, and invasion.''; PubMed Europe PMC Scholia
  64. Bione S, Sala C, Manzini C, Arrigo G, Zuffardi O, Banfi S, Borsani G, Jonveaux P, Philippe C, Zuccotti M, Ballabio A, Toniolo D.; ''A human homologue of the Drosophila melanogaster diaphanous gene is disrupted in a patient with premature ovarian failure: evidence for conserved function in oogenesis and implications for human sterility.''; PubMed Europe PMC Scholia
  65. Zong H, Raman N, Mickelson-Young LA, Atkinson SJ, Quilliam LA.; ''Loop 6 of RhoA confers specificity for effector binding, stress fiber formation, and cellular transformation.''; PubMed Europe PMC Scholia
  66. Yamashiro S, Totsukawa G, Yamakita Y, Sasaki Y, Madaule P, Ishizaki T, Narumiya S, Matsumura F.; ''Citron kinase, a Rho-dependent kinase, induces di-phosphorylation of regulatory light chain of myosin II.''; PubMed Europe PMC Scholia
  67. Roskoski R.; ''MEK1/2 dual-specificity protein kinases: structure and regulation.''; PubMed Europe PMC Scholia
  68. Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, O'Keefe CL, Matera AG, Xiong Y.; ''Growth suppression by p18, a p16INK4/MTS1- and p14INK4B/MTS2-related CDK6 inhibitor, correlates with wild-type pRb function.''; PubMed Europe PMC Scholia
  69. Torbett NE, Casamassima A, Parker PJ.; ''Hyperosmotic-induced protein kinase N 1 activation in a vesicular compartment is dependent upon Rac1 and 3-phosphoinositide-dependent kinase 1.''; PubMed Europe PMC Scholia
  70. Cseh B, Doma E, Baccarini M.; ''"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway.''; PubMed Europe PMC Scholia
  71. Fukata M, Kuroda S, Fujii K, Nakamura T, Shoji I, Matsuura Y, Okawa K, Iwamatsu A, Kikuchi A, Kaibuchi K.; ''Regulation of cross-linking of actin filament by IQGAP1, a target for Cdc42.''; PubMed Europe PMC Scholia
  72. Regan Anderson TM, Ma SH, Raj GV, Cidlowski JA, Helle TM, Knutson TP, Krutilina RI, Seagroves TN, Lange CA.; ''Breast Tumor Kinase (Brk/PTK6) Is Induced by HIF, Glucocorticoid Receptor, and PELP1-Mediated Stress Signaling in Triple-Negative Breast Cancer.''; PubMed Europe PMC Scholia
  73. Fernandez-Borja M, Janssen L, Verwoerd D, Hordijk P, Neefjes J.; ''RhoB regulates endosome transport by promoting actin assembly on endosomal membranes through Dia1.''; PubMed Europe PMC Scholia
  74. Ikeda O, Miyasaka Y, Sekine Y, Mizushima A, Muromoto R, Nanbo A, Yoshimura A, Matsuda T.; ''STAP-2 is phosphorylated at tyrosine-250 by Brk and modulates Brk-mediated STAT3 activation.''; PubMed Europe PMC Scholia
  75. Bashour AM, Fullerton AT, Hart MJ, Bloom GS.; ''IQGAP1, a Rac- and Cdc42-binding protein, directly binds and cross-links microfilaments.''; PubMed Europe PMC Scholia
  76. Miah S, Goel RK, Dai C, Kalra N, Beaton-Brown E, Bagu ET, Bonham K, Lukong KE.; ''BRK targets Dok1 for ubiquitin-mediated proteasomal degradation to promote cell proliferation and migration.''; PubMed Europe PMC Scholia
  77. Chastkofsky MI, Bie W, Ball-Kell SM, He YY, Tyner AL.; ''Protein Tyrosine Kinase 6 Regulates UVB-Induced Signaling and Tumorigenesis in Mouse Skin.''; PubMed Europe PMC Scholia
  78. Zheng Y, Peng M, Wang Z, Asara JM, Tyner AL.; ''Protein tyrosine kinase 6 directly phosphorylates AKT and promotes AKT activation in response to epidermal growth factor.''; PubMed Europe PMC Scholia
  79. Daniels RH, Bokoch GM.; ''p21-activated protein kinase: a crucial component of morphological signaling?''; PubMed Europe PMC Scholia
  80. Modha R, Campbell LJ, Nietlispach D, Buhecha HR, Owen D, Mott HR.; ''The Rac1 polybasic region is required for interaction with its effector PRK1.''; PubMed Europe PMC Scholia
  81. Misaki K, Mukai H, Yoshinaga C, Oishi K, Isagawa T, Takahashi M, Ohsumi K, Kishimoto T, Ono Y.; ''PKN delays mitotic timing by inhibition of Cdc25C: possible involvement of PKN in the regulation of cell division.''; PubMed Europe PMC Scholia
  82. Ono H, Basson MD, Ito H.; ''PTK6 promotes cancer migration and invasion in pancreatic cancer cells dependent on ERK signaling.''; PubMed Europe PMC Scholia
  83. Manser E, Leung T, Salihuddin H, Zhao ZS, Lim L.; ''A brain serine/threonine protein kinase activated by Cdc42 and Rac1.''; PubMed Europe PMC Scholia
  84. Kühn S, Geyer M.; ''Formins as effector proteins of Rho GTPases.''; PubMed Europe PMC Scholia
  85. Parrini MC, Lei M, Harrison SC, Mayer BJ.; ''Pak1 kinase homodimers are autoinhibited in trans and dissociated upon activation by Cdc42 and Rac1.''; PubMed Europe PMC Scholia
  86. Minoguchi M, Minoguchi S, Aki D, Joo A, Yamamoto T, Yumioka T, Matsuda T, Yoshimura A.; ''STAP-2/BKS, an adaptor/docking protein, modulates STAT3 activation in acute-phase response through its YXXQ motif.''; PubMed Europe PMC Scholia
  87. Hiebert SW.; ''Regions of the retinoblastoma gene product required for its interaction with the E2F transcription factor are necessary for E2 promoter repression and pRb-mediated growth suppression.''; PubMed Europe PMC Scholia
  88. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA.; ''Mutations of the BRAF gene in human cancer.''; PubMed Europe PMC Scholia
  89. Kovar DR, Harris ES, Mahaffy R, Higgs HN, Pollard TD.; ''Control of the assembly of ATP- and ADP-actin by formins and profilin.''; PubMed Europe PMC Scholia
  90. Hamaguchi T, Ito M, Feng J, Seko T, Koyama M, Machida H, Takase K, Amano M, Kaibuchi K, Hartshorne DJ, Nakano T.; ''Phosphorylation of CPI-17, an inhibitor of myosin phosphatase, by protein kinase N.''; PubMed Europe PMC Scholia
  91. Isobe A, Takeda T, Sakata M, Yamamoto T, Minekawa R, Hayashi M, Auernhammer CJ, Tasaka K, Murata Y.; ''STAT3-mediated constitutive expression of SOCS3 in an undifferentiated rat trophoblast-like cell line.''; PubMed Europe PMC Scholia
  92. Chakraborty G, Jain S, Kundu GC.; ''Osteopontin promotes vascular endothelial growth factor-dependent breast tumor growth and angiogenesis via autocrine and paracrine mechanisms.''; PubMed Europe PMC Scholia
  93. Colón-Franco JM, Gomez TS, Billadeau DD.; ''Dynamic remodeling of the actin cytoskeleton by FMNL1γ is required for structural maintenance of the Golgi complex.''; PubMed Europe PMC Scholia
  94. Fan L, Pellegrin S, Scott A, Mellor H.; ''The small GTPase Rif is an alternative trigger for the formation of actin stress fibers in epithelial cells.''; PubMed Europe PMC Scholia
  95. Miralles F, Posern G, Zaromytidou AI, Treisman R.; ''Actin dynamics control SRF activity by regulation of its coactivator MAL.''; PubMed Europe PMC Scholia
  96. Matsuzawa K, Kosako H, Inagaki N, Shibata H, Mukai H, Ono Y, Amano M, Kaibuchi K, Matsuura Y, Azuma I, Inagaki M.; ''Domain-specific phosphorylation of vimentin and glial fibrillary acidic protein by PKN.''; PubMed Europe PMC Scholia
  97. Wang J, Wu JW, Wang ZX.; ''Mechanistic studies of the autoactivation of PAK2: a two-step model of cis initiation followed by trans amplification.''; PubMed Europe PMC Scholia
  98. Di Cunto F, Calautti E, Hsiao J, Ong L, Topley G, Turco E, Dotto GP.; ''Citron rho-interacting kinase, a novel tissue-specific ser/thr kinase encompassing the Rho-Rac-binding protein Citron.''; PubMed Europe PMC Scholia
  99. Zhang B, Chernoff J, Zheng Y.; ''Interaction of Rac1 with GTPase-activating proteins and putative effectors. A comparison with Cdc42 and RhoA.''; PubMed Europe PMC Scholia
  100. Ikeda O, Mizushima A, Sekine Y, Yamamoto C, Muromoto R, Nanbo A, Oritani K, Yoshimura A, Matsuda T.; ''Involvement of STAP-2 in Brk-mediated phosphorylation and activation of STAT5 in breast cancer cells.''; PubMed Europe PMC Scholia
  101. Tanizaki J, Okamoto I, Sakai K, Nakagawa K.; ''Differential roles of trans-phosphorylated EGFR, HER2, HER3, and RET as heterodimerisation partners of MET in lung cancer with MET amplification.''; PubMed Europe PMC Scholia
  102. Li F, Higgs HN.; ''The mouse Formin mDia1 is a potent actin nucleation factor regulated by autoinhibition.''; PubMed Europe PMC Scholia
  103. Peng M, Ball-Kell SM, Tyner AL.; ''Protein tyrosine kinase 6 promotes ERBB2-induced mammary gland tumorigenesis in the mouse.''; PubMed Europe PMC Scholia
  104. Tominaga T, Sahai E, Chardin P, McCormick F, Courtneidge SA, Alberts AS.; ''Diaphanous-related formins bridge Rho GTPase and Src tyrosine kinase signaling.''; PubMed Europe PMC Scholia
  105. McKay MM, Morrison DK.; ''Integrating signals from RTKs to ERK/MAPK.''; PubMed Europe PMC Scholia
  106. Nizet V, Johnson RS.; ''Interdependence of hypoxic and innate immune responses.''; PubMed Europe PMC Scholia
  107. Xiang B, Chatti K, Qiu H, Lakshmi B, Krasnitz A, Hicks J, Yu M, Miller WT, Muthuswamy SK.; ''Brk is coamplified with ErbB2 to promote proliferation in breast cancer.''; PubMed Europe PMC Scholia
  108. Holinstat M, Knezevic N, Broman M, Samarel AM, Malik AB, Mehta D.; ''Suppression of RhoA activity by focal adhesion kinase-induced activation of p190RhoGAP: role in regulation of endothelial permeability.''; PubMed Europe PMC Scholia
  109. King PD, Lubeck BA, Lapinski PE.; ''Nonredundant functions for Ras GTPase-activating proteins in tissue homeostasis.''; PubMed Europe PMC Scholia
  110. Zheng Y, Asara JM, Tyner AL.; ''Protein-tyrosine kinase 6 promotes peripheral adhesion complex formation and cell migration by phosphorylating p130 CRK-associated substrate.''; PubMed Europe PMC Scholia
  111. Kato T, Gotoh Y, Hoffmann A, Ono Y.; ''Negative regulation of constitutive NF-kappaB and JNK signaling by PKN1-mediated phosphorylation of TRAF1.''; PubMed Europe PMC Scholia
  112. Serrano M, Hannon GJ, Beach D.; ''A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4.''; PubMed Europe PMC Scholia
  113. Sadasivam S, DeCaprio JA.; ''The DREAM complex: master coordinator of cell cycle-dependent gene expression.''; PubMed Europe PMC Scholia
  114. Manser E, Chong C, Zhao ZS, Leung T, Michael G, Hall C, Lim L.; ''Molecular cloning of a new member of the p21-Cdc42/Rac-activated kinase (PAK) family.''; PubMed Europe PMC Scholia
  115. Chen HY, Shen CH, Tsai YT, Lin FC, Huang YP, Chen RH.; ''Brk activates rac1 and promotes cell migration and invasion by phosphorylating paxillin.''; PubMed Europe PMC Scholia
  116. Camera P, da Silva JS, Griffiths G, Giuffrida MG, Ferrara L, Schubert V, Imarisio S, Silengo L, Dotti CG, Di Cunto F.; ''Citron-N is a neuronal Rho-associated protein involved in Golgi organization through actin cytoskeleton regulation.''; PubMed Europe PMC Scholia
  117. Cheng M, Sexl V, Sherr CJ, Roussel MF.; ''Assembly of cyclin D-dependent kinase and titration of p27Kip1 regulated by mitogen-activated protein kinase kinase (MEK1).''; PubMed Europe PMC Scholia
  118. Cheng L, Zhang J, Ahmad S, Rozier L, Yu H, Deng H, Mao Y.; ''Aurora B regulates formin mDia3 in achieving metaphase chromosome alignment.''; PubMed Europe PMC Scholia
  119. Kaelin WG, Ratcliffe PJ.; ''Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway.''; PubMed Europe PMC Scholia
  120. Kang SA, Lee ST.; ''PTK6 promotes degradation of c-Cbl through PTK6-mediated phosphorylation.''; PubMed Europe PMC Scholia
  121. Tian YM, Yeoh KK, Lee MK, Eriksson T, Kessler BM, Kramer HB, Edelmann MJ, Willam C, Pugh CW, Schofield CJ, Ratcliffe PJ.; ''Differential sensitivity of hypoxia inducible factor hydroxylation sites to hypoxia and hydroxylase inhibitors.''; PubMed Europe PMC Scholia
  122. Plotnikov A, Zehorai E, Procaccia S, Seger R.; ''The MAPK cascades: signaling components, nuclear roles and mechanisms of nuclear translocation.''; PubMed Europe PMC Scholia
  123. Lukong KE, Richard S.; ''Breast tumor kinase BRK requires kinesin-2 subunit KAP3A in modulation of cell migration.''; PubMed Europe PMC Scholia
  124. Chong C, Tan L, Lim L, Manser E.; ''The mechanism of PAK activation. Autophosphorylation events in both regulatory and kinase domains control activity.''; PubMed Europe PMC Scholia
  125. Semenza GL.; ''Life with oxygen.''; PubMed Europe PMC Scholia
  126. Kitzing TM, Wang Y, Pertz O, Copeland JW, Grosse R.; ''Formin-like 2 drives amoeboid invasive cell motility downstream of RhoC.''; PubMed Europe PMC Scholia
  127. Swart-Mataraza JM, Li Z, Sacks DB.; ''IQGAP1 is a component of Cdc42 signaling to the cytoskeleton.''; PubMed Europe PMC Scholia
  128. Ferreira R, Magnaghi-Jaulin L, Robin P, Harel-Bellan A, Trouche D.; ''The three members of the pocket proteins family share the ability to repress E2F activity through recruitment of a histone deacetylase.''; PubMed Europe PMC Scholia
  129. Gasman S, Kalaidzidis Y, Zerial M.; ''RhoD regulates endosome dynamics through Diaphanous-related Formin and Src tyrosine kinase.''; PubMed Europe PMC Scholia
  130. Yayoshi-Yamamoto S, Taniuchi I, Watanabe T.; ''FRL, a novel formin-related protein, binds to Rac and regulates cell motility and survival of macrophages.''; PubMed Europe PMC Scholia
  131. Vidal A, Koff A.; ''Cell-cycle inhibitors: three families united by a common cause.''; PubMed Europe PMC Scholia
  132. Lei M, Lu W, Meng W, Parrini MC, Eck MJ, Mayer BJ, Harrison SC.; ''Structure of PAK1 in an autoinhibited conformation reveals a multistage activation switch.''; PubMed Europe PMC Scholia
  133. Connell-Crowley L, Harper JW, Goodrich DW.; ''Cyclin D1/Cdk4 regulates retinoblastoma protein-mediated cell cycle arrest by site-specific phosphorylation.''; PubMed Europe PMC Scholia
  134. Coyle JH, Guzik BW, Bor YC, Jin L, Eisner-Smerage L, Taylor SJ, Rekosh D, Hammarskjöld ML.; ''Sam68 enhances the cytoplasmic utilization of intron-containing RNA and is functionally regulated by the nuclear kinase Sik/BRK.''; PubMed Europe PMC Scholia
  135. Nezami AG, Poy F, Eck MJ.; ''Structure of the autoinhibitory switch in formin mDia1.''; PubMed Europe PMC Scholia
  136. Liu L, Gao Y, Qiu H, Miller WT, Poli V, Reich NC.; ''Identification of STAT3 as a specific substrate of breast tumor kinase.''; PubMed Europe PMC Scholia
  137. Wallar BJ, Deward AD, Resau JH, Alberts AS.; ''RhoB and the mammalian Diaphanous-related formin mDia2 in endosome trafficking.''; PubMed Europe PMC Scholia
  138. Zhang H.; ''Life without kinase: cyclin E promotes DNA replication licensing and beyond.''; PubMed Europe PMC Scholia
  139. Brahimi-Horn MC, Pouysségur J.; ''HIF at a glance.''; PubMed Europe PMC Scholia
  140. Lane J, Martin T, Weeks HP, Jiang WG.; ''Structure and role of WASP and WAVE in Rho GTPase signalling in cancer.''; PubMed Europe PMC Scholia
  141. Jung JH, Traugh JA.; ''Regulation of the interaction of Pak2 with Cdc42 via autophosphorylation of serine 141.''; PubMed Europe PMC Scholia
  142. Haegebarth A, Heap D, Bie W, Derry JJ, Richard S, Tyner AL.; ''The nuclear tyrosine kinase BRK/Sik phosphorylates and inhibits the RNA-binding activities of the Sam68-like mammalian proteins SLM-1 and SLM-2.''; PubMed Europe PMC Scholia
  143. Roberts PJ, Der CJ.; ''Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer.''; PubMed Europe PMC Scholia
  144. Wu L, Timmers C, Maiti B, Saavedra HI, Sang L, Chong GT, Nuckolls F, Giangrande P, Wright FA, Field SJ, Greenberg ME, Orkin S, Nevins JR, Robinson ML, Leone G.; ''The E2F1-3 transcription factors are essential for cellular proliferation.''; PubMed Europe PMC Scholia
  145. Parry D, Bates S, Mann DJ, Peters G.; ''Lack of cyclin D-Cdk complexes in Rb-negative cells correlates with high levels of p16INK4/MTS1 tumour suppressor gene product.''; PubMed Europe PMC Scholia
  146. Wells A.; ''EGF receptor.''; PubMed Europe PMC Scholia
  147. Turjanski AG, Vaqué JP, Gutkind JS.; ''MAP kinases and the control of nuclear events.''; PubMed Europe PMC Scholia
  148. Wiejak J, Dunlop J, Gao S, Borland G, Yarwood SJ.; ''Extracellular signal-regulated kinase mitogen-activated protein kinase-dependent SOCS-3 gene induction requires c-Jun, signal transducer and activator of transcription 3, and specificity protein 3 transcription factors.''; PubMed Europe PMC Scholia
  149. Brill S, Li S, Lyman CW, Church DM, Wasmuth JJ, Weissbach L, Bernards A, Snijders AJ.; ''The Ras GTPase-activating-protein-related human protein IQGAP2 harbors a potential actin binding domain and interacts with calmodulin and Rho family GTPases.''; PubMed Europe PMC Scholia
  150. Semenza GL.; ''Hydroxylation of HIF-1: oxygen sensing at the molecular level.''; PubMed Europe PMC Scholia
  151. Ahmadian MR, Kiel C, Stege P, Scheffzek K.; ''Structural fingerprints of the Ras-GTPase activating proteins neurofibromin and p120GAP.''; PubMed Europe PMC Scholia
  152. Bassi ZI, Audusseau M, Riparbelli MG, Callaini G, D'Avino PP.; ''Citron kinase controls a molecular network required for midbody formation in cytokinesis.''; PubMed Europe PMC Scholia
  153. Hannon GJ, Beach D.; ''p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest.''; PubMed Europe PMC Scholia
  154. Wellbrock C, Karasarides M, Marais R.; ''The RAF proteins take centre stage.''; PubMed Europe PMC Scholia
  155. Brown MD, Sacks DB.; ''Protein scaffolds in MAP kinase signalling.''; PubMed Europe PMC Scholia
  156. Collazos A, Michael N, Whelan RD, Kelly G, Mellor H, Pang LC, Totty N, Parker PJ.; ''Site recognition and substrate screens for PKN family proteins.''; PubMed Europe PMC Scholia
  157. Wang S, Watanabe T, Noritake J, Fukata M, Yoshimura T, Itoh N, Harada T, Nakagawa M, Matsuura Y, Arimura N, Kaibuchi K.; ''IQGAP3, a novel effector of Rac1 and Cdc42, regulates neurite outgrowth.''; PubMed Europe PMC Scholia
  158. Lammers M, Rose R, Scrima A, Wittinghofer A.; ''The regulation of mDia1 by autoinhibition and its release by Rho*GTP.''; PubMed Europe PMC Scholia
  159. Vega FM, Fruhwirth G, Ng T, Ridley AJ.; ''RhoA and RhoC have distinct roles in migration and invasion by acting through different targets.''; PubMed Europe PMC Scholia
  160. Scheffzek K, Ahmadian MR, Kabsch W, Wiesmüller L, Lautwein A, Schmitz F, Wittinghofer A.; ''The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants.''; PubMed Europe PMC Scholia
  161. Pasterkamp RJ, Kolodkin AL.; ''Semaphorin junction: making tracks toward neural connectivity.''; PubMed Europe PMC Scholia
  162. Fan G, Lin G, Lucito R, Tonks NK.; ''Protein-tyrosine phosphatase 1B antagonized signaling by insulin-like growth factor-1 receptor and kinase BRK/PTK6 in ovarian cancer cells.''; PubMed Europe PMC Scholia
  163. Ikeda O, Sekine Y, Mizushima A, Nakasuji M, Miyasaka Y, Yamamoto C, Muromoto R, Nanbo A, Oritani K, Yoshimura A, Matsuda T.; ''Interactions of STAP-2 with Brk and STAT3 participate in cell growth of human breast cancer cells.''; PubMed Europe PMC Scholia
  164. Schlessinger J.; ''Ligand-induced, receptor-mediated dimerization and activation of EGF receptor.''; PubMed Europe PMC Scholia
  165. Zhang W, Benson DL.; ''Targeting and clustering citron to synapses.''; PubMed Europe PMC Scholia
  166. Yang J, Huang J, Dasgupta M, Sears N, Miyagi M, Wang B, Chance MR, Chen X, Du Y, Wang Y, An L, Wang Q, Lu T, Zhang X, Wang Z, Stark GR.; ''Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes.''; PubMed Europe PMC Scholia
  167. Bagchi S, Weinmann R, Raychaudhuri P.; ''The retinoblastoma protein copurifies with E2F-I, an E1A-regulated inhibitor of the transcription factor E2F.''; PubMed Europe PMC Scholia
  168. Szczepanowska J.; ''Involvement of Rac/Cdc42/PAK pathway in cytoskeletal rearrangements.''; PubMed Europe PMC Scholia
  169. Guan KL, Jenkins CW, Li Y, O'Keefe CL, Noh S, Wu X, Zariwala M, Matera AG, Xiong Y.; ''Isolation and characterization of p19INK4d, a p16-related inhibitor specific to CDK6 and CDK4.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114927view16:44, 25 January 2021ReactomeTeamReactome version 75
113372view11:44, 2 November 2020ReactomeTeamReactome version 74
112576view15:54, 9 October 2020ReactomeTeamReactome version 73
102035view16:22, 26 November 2018Marvin M2Ontology Term : 'PW:0000003' removed !
102034view16:21, 26 November 2018Marvin M2Ontology Term : 'kinase mediated signaling pathway' added !
101698view14:35, 1 November 2018DeSlOntology Term : 'signaling pathway' added !
101490view11:35, 1 November 2018ReactomeTeamreactome version 66
101027view21:15, 31 October 2018ReactomeTeamreactome version 65
100725view20:11, 31 October 2018ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
11DCORST MetaboliteCHEBI:16973 (ChEBI)
ADPMetaboliteCHEBI:456216 (ChEBI)
AKT1 ProteinP31749 (Uniprot-TrEMBL)
AKT1ProteinP31749 (Uniprot-TrEMBL)
ALDO MetaboliteCHEBI:27584 (ChEBI)
ARAP1 ProteinQ96P48 (Uniprot-TrEMBL)
ARAP1ProteinQ96P48 (Uniprot-TrEMBL)
ARHGAP35 ProteinQ9NRY4 (Uniprot-TrEMBL)
ARHGAP35ProteinQ9NRY4 (Uniprot-TrEMBL)
ATPMetaboliteCHEBI:30616 (ChEBI)
Autophosphorylated p-Y877-ERBB2 heterodimers R-HSA-1963580 (Reactome)
BCAR1 ProteinP56945 (Uniprot-TrEMBL)
BCAR1ProteinP56945 (Uniprot-TrEMBL)
CBLProteinP22681 (Uniprot-TrEMBL)
CCND1 ProteinP24385 (Uniprot-TrEMBL)
CCNE1 ProteinP24864 (Uniprot-TrEMBL)
CDK2 ProteinP24941 (Uniprot-TrEMBL)
CDK4 ProteinP11802 (Uniprot-TrEMBL)
CDKN1B ProteinP46527 (Uniprot-TrEMBL)
CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))ComplexR-HSA-8848419 (Reactome)
CORST MetaboliteCHEBI:16827 (ChEBI)
CORT MetaboliteCHEBI:17650 (ChEBI)
CRK ProteinP46108 (Uniprot-TrEMBL)
CRK:DOCK180:ELMO1,ELMO2ComplexR-HSA-2029141 (Reactome)
Cellular response to hypoxiaPathwayR-HSA-1234174 (Reactome) Oxygen plays a central role in the functioning of human cells: it is both essential for normal metabolism and toxic. Here we have annotated one aspect of cellular responses to oxygen, the role of hypoxia-inducible factor in regulating cellular transcriptional responses to changes in oxygen availability.

In the presence of oxygen members of the transcription factor family HIF-alpha, comprising HIF1A, HIF2A (EPAS1), and HIF3A, are hydroxylated on proline residues by PHD1 (EGLN2), PHD2 (EGLN1), and PHD3 (EGLN3) and on asparagine residues by HIF1AN (FIH) (reviewed in Pouyssegur et al. 2006, Semenza 2007, Kaelin and Ratcliffe 2008, Nizet and Johnson 2009, Brahimi-Horn and Pouyssegur 2009, Majmundar et al. 2010, Loenarz and Schofield 2011). Both types of reaction require molecular oxygen as a substrate and it is probable that at least some HIF-alpha molecules carry both hydroxylated asparagine and hydroxylated proline (Tian et al. 2011).
Hydroxylated asparagine interferes with the ability of HIF-alpha to interact with p300 and CBP while hydroxylated proline facilitates the interaction of HIF-alpha with the E3 ubiquitin ligase VHL, causing ubiquitination and proteolysis of HIF-alpha. Hypoxia inhibits both types of hydroxylation, resulting in the stabilization of HIF-alpha, which then enters the nucleus, binds HIF-beta, and recruits p300 and CBP to activate target genes such as EPO and VEGF.

DOCK1 ProteinQ14185 (Uniprot-TrEMBL)
DOK1 ProteinQ99704 (Uniprot-TrEMBL)
DOK1ProteinQ99704 (Uniprot-TrEMBL)
EGFR ProteinP00533 (Uniprot-TrEMBL)
ELMO1 ProteinQ92556 (Uniprot-TrEMBL)
ELMO2 ProteinQ96JJ3 (Uniprot-TrEMBL)
EPAS1 ProteinQ99814 (Uniprot-TrEMBL)
EPAS1ProteinQ99814 (Uniprot-TrEMBL)
GDP MetaboliteCHEBI:17552 (ChEBI)
GDPMetaboliteCHEBI:17552 (ChEBI)
GPNMB ProteinQ14956 (Uniprot-TrEMBL)
GPNMBProteinQ14956 (Uniprot-TrEMBL)
GTP MetaboliteCHEBI:15996 (ChEBI)
GTPMetaboliteCHEBI:15996 (ChEBI)
H2OMetaboliteCHEBI:15377 (ChEBI)
HBEGF(63-148) ProteinQ99075 (Uniprot-TrEMBL)
HBEGF:EGFR:GPNMBComplexR-HSA-8857546 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:PTK6:LRRK2ComplexR-HSA-8857567 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:p-Y351-PTK6:LRRK2ComplexR-HSA-8857574 (Reactome)
HBEGF:EGFR:p-Y525-GPNMBComplexR-HSA-8857556 (Reactome)
HBEGF:EGFRComplexR-HSA-8857547 (Reactome)
HIF1A ProteinQ16665 (Uniprot-TrEMBL)
HIF1A,EPAS1:PTK6 GeneComplexR-HSA-8848808 (Reactome)
HIF1A,EPAS1ComplexR-HSA-8848802 (Reactome)
HIF1AProteinQ16665 (Uniprot-TrEMBL)
KHDRBS1 ProteinQ07666 (Uniprot-TrEMBL)
KHDRBS1ProteinQ07666 (Uniprot-TrEMBL)
KHDRBS2ProteinQ5VWX1 (Uniprot-TrEMBL)
KHDRSB3ProteinO75525 (Uniprot-TrEMBL)
LINC01139 ProteinENST00000400946 (Ensembl)
LINC01139RnaENST00000400946 (Ensembl)
LRRK2 ProteinQ5S007 (Uniprot-TrEMBL)
LRRK2ProteinQ5S007 (Uniprot-TrEMBL)
Mitotic G1 phase and G1/S transitionPathwayR-HSA-453279 (Reactome) Mitotic G1-G1/S phase involves G1 phase of the mitotic interphase and G1/S transition, when a cell commits to DNA replication and divison genetic and cellular material to two daughter cells.

During early G1, cells can enter a quiescent G0 state. In quiescent cells, the evolutionarily conserved DREAM complex, consisting of the pocket protein family member p130 (RBL2), bound to E2F4 or E2F5, and the MuvB complex, represses transcription of cell cycle genes (reviewed by Sadasivam and DeCaprio 2013).

During early G1 phase in actively cycling cells, transcription of cell cycle genes is repressed by another pocket protein family member, p107 (RBL1), which forms a complex with E2F4 (Ferreira et al. 1998, Cobrinik 2005). RB1 tumor suppressor, the product of the retinoblastoma susceptibility gene, is the third member of the pocket protein family. RB1 binds to E2F transcription factors E2F1, E2F2 and E2F3 and inhibits their transcriptional activity, resulting in prevention of G1/S transition (Chellappan et al. 1991, Bagchi et al. 1991, Chittenden et al. 1991, Lees et al. 1993, Hiebert 1993, Wu et al. 2001). Once RB1 is phosphorylated on serine residue S795 by Cyclin D:CDK4/6 complexes, it can no longer associate with and inhibit E2F1-3. Thus, CDK4/6-mediated phosphorylation of RB1 leads to transcriptional activation of E2F1-3 target genes needed for the S phase of the cell cycle (Connell-Crowley et al. 1997). CDK2, in complex with cyclin E, contributes to RB1 inactivation and also activates proteins needed for the initiation of DNA replication (Zhang 2007). Expression of D type cyclins is regulated by extracellular mitogens (Cheng et al. 1998, Depoortere et al. 1998). Catalytic activities of CDK4/6 and CDK2 are controlled by CDK inhibitors of the INK4 family (Serrano et al. 1993, Hannon and Beach 1994, Guan et al. 1994, Guan et al. 1996, Parry et al. 1995) and the Cip/Kip family, respectively.

NR3C1 ProteinP04150 (Uniprot-TrEMBL)
NR3C1:(ALDO,11DCORST,CORST,CORT) dimerComplexR-HSA-879850 (Reactome)
PELP1 ProteinQ8IZL8 (Uniprot-TrEMBL)
PELP1ProteinQ8IZL8 (Uniprot-TrEMBL)
PIP3 activates AKT signalingPathwayR-HSA-1257604 (Reactome) Signaling by AKT is one of the key outcomes of receptor tyrosine kinase (RTK) activation. AKT is activated by the cellular second messenger PIP3, a phospholipid that is generated by PI3K. In ustimulated cells, PI3K class IA enzymes reside in the cytosol as inactive heterodimers composed of p85 regulatory subunit and p110 catalytic subunit. In this complex, p85 stabilizes p110 while inhibiting its catalytic activity. Upon binding of extracellular ligands to RTKs, receptors dimerize and undergo autophosphorylation. The regulatory subunit of PI3K, p85, is recruited to phosphorylated cytosolic RTK domains either directly or indirectly, through adaptor proteins, leading to a conformational change in the PI3K IA heterodimer that relieves inhibition of the p110 catalytic subunit. Activated PI3K IA phosphorylates PIP2, converting it to PIP3; this reaction is negatively regulated by PTEN phosphatase. PIP3 recruits AKT to the plasma membrane, allowing TORC2 to phosphorylate a conserved serine residue of AKT. Phosphorylation of this serine induces a conformation change in AKT, exposing a conserved threonine residue that is then phosphorylated by PDPK1 (PDK1). Phosphorylation of both the threonine and the serine residue is required to fully activate AKT. The active AKT then dissociates from PIP3 and phosphorylates a number of cytosolic and nuclear proteins that play important roles in cell survival and metabolism. For a recent review of AKT signaling, please refer to Manning and Cantley, 2007.
PTK6 Gene:EPAS1:NR3C1:Glucocorticoid ligand:PELP1ComplexR-HSA-8856931 (Reactome)
PTK6 Gene ProteinENSG00000101213 (Ensembl)
PTK6 GeneGeneProductENSG00000101213 (Ensembl)
PTK6 ProteinQ13882 (Uniprot-TrEMBL)
PTK6ProteinQ13882 (Uniprot-TrEMBL)
PTPN1 ProteinP18031 (Uniprot-TrEMBL)
PTPN1ProteinP18031 (Uniprot-TrEMBL)
PXN ProteinP49023 (Uniprot-TrEMBL)
PXNProteinP49023 (Uniprot-TrEMBL)
PiMetaboliteCHEBI:43474 (ChEBI)
PolyUb,p-Y700,Y731,Y774-CBLProteinP22681 (Uniprot-TrEMBL)
RAC1 ProteinP63000 (Uniprot-TrEMBL)
RAC1:GDPComplexR-HSA-5674631 (Reactome)
RAC1:GTPComplexR-HSA-442641 (Reactome)
RAF/MAP kinase cascadePathwayR-HSA-5673001 (Reactome) The RAS-RAF-MEK-ERK pathway regulates processes such as proliferation, differentiation, survival, senescence and cell motility in response to growth factors, hormones and cytokines, among others. Binding of these stimuli to receptors in the plasma membrane promotes the GEF-mediated activation of RAS at the plasma membrane and initiates the three-tiered kinase cascade of the conventional MAPK cascades. GTP-bound RAS recruits RAF (the MAPK kinase kinase), and promotes its dimerization and activation (reviewed in Cseh et al, 2014; Roskoski, 2010; McKay and Morrison, 2007; Wellbrock et al, 2004). Activated RAF phosphorylates the MAPK kinase proteins MEK1 and MEK2 (also known as MAP2K1 and MAP2K2), which in turn phophorylate the proline-directed kinases ERK1 and 2 (also known as MAPK3 and MAPK1) (reviewed in Roskoski, 2012a, b; Kryiakis and Avruch, 2012). Activated ERK proteins may undergo dimerization and have identified targets in both the nucleus and the cytosol; consistent with this, a proportion of activated ERK protein relocalizes to the nucleus in response to stimuli (reviewed in Roskoski 2012b; Turjanski et al, 2007; Plotnikov et al, 2010; Cargnello et al, 2011). Although initially seen as a linear cascade originating at the plasma membrane and culminating in the nucleus, the RAS/RAF MAPK cascade is now also known to be activated from various intracellular location. Temporal and spatial specificity of the cascade is achieved in part through the interaction of pathway components with numerous scaffolding proteins (reviewed in McKay and Morrison, 2007; Brown and Sacks, 2009).
The importance of the RAS/RAF MAPK cascade is highlighted by the fact that components of this pathway are mutated with high frequency in a large number of human cancers. Activating mutations in RAS are found in approximately one third of human cancers, while ~8% of tumors express an activated form of BRAF (Roberts and Der, 2007; Davies et al, 2002; Cantwell-Dorris et al, 2011).
RASA1 ProteinP20936 (Uniprot-TrEMBL)
RASA1ProteinP20936 (Uniprot-TrEMBL)
RHO GTPases Activate ForminsPathwayR-HSA-5663220 (Reactome) Formins are a family of proteins with 15 members in mammals, organized into 8 subfamilies. Formins are involved in the regulation of actin cytoskeleton. Many but not all formin family members are activated by RHO GTPases. Formins that serve as effectors of RHO GTPases belong to different formin subfamilies but they all share a structural similarity to Drosophila protein diaphanous and are hence named diaphanous-related formins (DRFs).

DRFs activated by RHO GTPases contain a GTPase binding domain (GBD) at their N-terminus, followed by formin homology domains 3, 1, and 2 (FH3, FH1, FH2) and a diaphanous autoregulatory domain (DAD) at the C-terminus. Most DRFs contain a dimerization domain (DD) and a coiled-coil region (CC) in between FH3 and FH1 domains (reviewed by Kuhn and Geyer 2014). RHO GTPase-activated DRFs are autoinhibited through the interaction between FH3 and DAD which is disrupted upon binding to an active RHO GTPase (Li and Higgs 2003, Lammers et al. 2005, Nezami et al. 2006). Since formins dimerize, it is not clear whether the FH3-DAD interaction is intra- or intermolecular. FH2 domain is responsible for binding to the F-actin and contributes to the formation of head-to-tail formin dimers (Xu et al. 2004). The proline-rich FH1 domain interacts with the actin-binding proteins profilins, thereby facilitating actin recruitment to formins and accelerating actin polymerization (Romero et al. 2004, Kovar et al. 2006).

Different formins are activated by different RHO GTPases in different cell contexts. FMNL1 (formin-like protein 1) is activated by binding to the RAC1:GTP and is involved in the formation of lamellipodia in macrophages (Yayoshi-Yamamoto et al. 2000) and is involved in the regulation of the Golgi complex structure (Colon-Franco et al. 2011). Activation of FMNL1 by CDC42:GTP contributes to the formation of the phagocytic cup (Seth et al. 2006). Activation of FMNL2 (formin-like protein 2) and FMNL3 (formin-like protein 3) by RHOC:GTP is involved in cancer cell motility and invasiveness (Kitzing et al. 2010, Vega et al. 2011). DIAPH1, activated by RHOA:GTP, promotes elongation of actin filaments and activation of SRF-mediated transcription which is inhibited by unpolymerized actin (Miralles et al. 2003). RHOF-mediated activation of DIAPH1 is implicated in formation of stress fibers (Fan et al. 2010). Activation of DIAPH1 and DIAPH3 by RHOB:GTP leads to actin coat formation around endosomes and regulates endosome motility and trafficking (Fernandez-Borja et al. 2005, Wallar et al. 2007). Endosome trafficking is also regulated by DIAPH2 transcription isoform 3 (DIAPH2-3) which, upon activation by RHOD:GTP, recruits SRC kinase to endosomes (Tominaga et al. 2000, Gasman et al. 2003). DIAPH2 transcription isoform 2 (DIAPH2-2) is involved in mitosis where, upon being activated by CDC42:GTP, it facilitates the capture of astral microtubules by kinetochores (Yasuda et al. 2004, Cheng et al. 2011). DIAPH2 is implicated in ovarian maintenance and premature ovarian failure (Bione et al. 1998). DAAM1, activated by RHOA:GTP, is involved in linking WNT signaling to cytoskeleton reorganization (Habas et al. 2001).

RHO GTPases Activate WASPs and WAVEsPathwayR-HSA-5663213 (Reactome) WASP and WAVE proteins belong to the Wiskott-Aldrich Syndrome protein family, with recessive mutations in the founding member WASP being responsible for the X-linked recessive immunodeficieny known as the Wiskott-Aldrich Syndrome. WASP proteins include WASP and WASL (N-WASP). WAVE proteins include WASF1 (WAVE1), WASF2 (WAVE2) and WASF3 (WAVE3). WASPs and WAVEs contain a VCA domain (consisting of WH2 and CA subdomains) at the C-terminus, responsible for binding to G-actin (WH2 subdomain) and the actin-associated ARP2/3 complex (CA subdomain). WASPs contain a WH1 (WASP homology 1) domain at the N-terminus, responsible for binding to WIPs (WASP-interacting proteins). A RHO GTPase binding domain (GBD) is located in the N-terminal half of WASPs and C-terminally located in WAVEs. RHO GTPases activate WASPs by disrupting the autoinhibitory interaction between the GBD and VCA domains, which allows WASPs to bind actin and the ARP2/3 complex and act as nucleation promoting factors in actin polymerization. WAVEs have the WAVE/SCAR homology domain (WHD/SHD) at the N-terminus, which binds ABI, NCKAP1, CYFIP2 and BRK1 to form the WAVE regulatory complex (WRC). Binding of the RAC1:GTP to the GBD of WAVEs most likely induces a conformational change in the WRC that allows activating phosphorylation of WAVEs by ABL1, thus enabling them to function as nucleation promoting factors in actin polymerization through binding G-actin and the ARP2/3 complex (Reviewed by Lane et al. 2014).
RHO GTPases activate CITPathwayR-HSA-5625900 (Reactome) Citron kinase (CIT) or citron RHO-interacting kinase (CRIK) shares similarities with ROCK kinases. Like ROCK, it consists of a serine/threonine kinase domain, a coiled-coil region, a RHO-binding domain, a cysteine rich region and a plekstrin homology (PH) domain, but additionally features a proline-rich region and a PDZ-binding domain. A shorter splicing isoform of CIT, citron-N, is specifically expressed in the nervous system and lacks the kinase domain. Citron-N is a component of the post-synaptic density, where it binds to the PDZ domains of the scaffolding protein PDS-95/SAP90 (Zhang et al. 2006).

While the binding of CIT to RHO GTPases RHOA, RHOB, RHOC and RAC1 is well established (Madaule et al. 1995), the mechanism of CIT activation by GTP-bound RHO GTPases has not been elucidated. There are indications that CIT may be activated through autophosphorylation in the presence of active forms of RHO GTPases (Di Cunto et al. 1998). CIT appears to phosphorylate the myosin regulatory light chain (MRLC), the only substrate identified to date, on the same residues that are phosphorylated by ROCKs, but it has not been established yet how this relates to activation by RHO GTPases (Yamashiro et al. 2003). CIT and RHOA are implicated to act together in Golgi apparatus organization through regulation of the actin cytoskeleton (Camera et al. 2003). CIT is also involved in the regulation of cytokinesis through its interaction with KIF14 (Gruneberg et al. 2006, Bassi et al. 2013, Watanabe et al. 2013) and p27(Kip1) (Serres et al. 2012).

RHO GTPases activate IQGAPsPathwayR-HSA-5626467 (Reactome) IQGAPs constitute a family of scaffolding proteins characterized by a calponin homology (CH) domain, a polyproline binding region (WW domain), a tandem of four IQ (isoleucine and glutamine-rich) repeats and a RAS GTPase-activating protein-related domain (GRD). Three IQGAPs have been identified in human, IQGAP1, IQGAP2 and IQGAP3. The best characterized is IQGAP1 and over 90 proteins have been reported to bind to it. IQGAPs integrate multiple signaling pathways and coordinate a large variety of cellular activities (White et al. 2012). IQGAP proteins IQGAP1, IQGAP2 and IQGAP3, bind activated RHO GTPases RAC1 and CDC42 via their GRD and stabilize them in their GTP-bound state (Kuroda et al. 1996, Swart-Mataraza et al. 2002, Wang et al. 2007). IQGAPs bind F-actin filaments via the CH domain and modulate cell shape and motility through regulation of G-actin/F-actin equilibrium (Brill et al. 1996, Fukata et al. 1997, Bashour et al. 1997, Wang et al. 2007, Pelikan-Conchaudron et al. 2011). Binding of IQGAPs to F-actin is inhibited by calmodulin binding to the IQ repeats (Bashour et al. 1997, Pelikan-Conchaudron et al. 2011). Based on IQGAP1 studies, IQGAPs presumably function as homodimers (Bashour et al. 1997).

IQGAP1 is involved in the regulation of adherens junctions through its interaction with E-cadherin (CDH1) and catenins (CTTNB1 and CTTNA1) (Kuroda et al. 1998, Hage et al. 2009). IQGAP1 contributes to cell polarity and lamellipodia formation through its interaction with microtubules (Fukata et al. 2002, Suzuki and Takahashi 2008).

RHO GTPases activate KTN1PathwayR-HSA-5625970 (Reactome) GTP-bound active forms of RHO GTPases RHOA, RHOG, RAC1 and CDC42 bind kinectin (KTN1), a protein inserted in endoplasmic reticulum membranes that interacts with the cargo-binding site of kinesin and activates its microtubule-stimulated ATPase activity required for vesicle motility (Vignal et al. 2001, Hotta et al. 1996). The effect of RHOG activity on cellular morphology, exhibited in the formation of microtubule-dependent cellular protrusions, depends both on RHOG interaction with KTN1, as well as on the kinesin activity (Vignal et al. 2001). RHOG and KTN1 also cooperate in microtubule-dependent lysosomal transport (Vignal et al. 2001). The precise mechanism of kinectin-mediated Rho GTPase signaling cascade needs further elucidation, and only the first two steps, KTN1-activated RHO GTPase binding, and KTN1-kinesin-1 binding are annotated here.
RHO GTPases activate PAKsPathwayR-HSA-5627123 (Reactome) The PAKs (p21-activated kinases) are a family of serine/threonine kinases mainly implicated in cytoskeletal rearrangements. All PAKs share a conserved catalytic domain located at the carboxyl terminus and a highly conserved motif in the amino terminus known as p21-binding domain (PBD) or Cdc42/Rac interactive binding (CRIB) domain. There are six mammalian PAKs that can be divided into two classes: class I (or conventional) PAKs (PAK1-3) and class II PAKs (PAK4-6). Conventional PAKs are important regulators of cytoskeletal dynamics and cell motility and are additionally implicated in transcription through MAPK (mitogen-activated protein kinase) cascades, death and survival signaling and cell cycle progression (Chan and Manser 2012).

PAK1, PAK2 and PAK3 are direct effectors of RAC1 and CDC42 GTPases. RAC1 and CDC42 bind to the CRIB domain. This binding induces a conformational change that disrupts inactive PAK homodimers and relieves autoinhibition of the catalytic carboxyl terminal domain (Manser et al. 1994, Manser et al. 1995, Zhang et al. 1998, Lei et al. 2000, Parrini et al. 2002; reviewed by Daniels and Bokoch 1999, Szczepanowska 2009). Autophosphorylation of a conserved threonine residue in the catalytic domain of PAKs (T423 in PAK1, T402 in PAK2 and T436 in PAK3) is necessary for the kinase activity of PAK1, PAK2 and PAK3. Autophosphorylation of PAK1 serine residue S144, PAK2 serine residue S141, and PAK3 serine residue S154 disrupts association of PAKs with RAC1 or CDC42 and enhances kinase activity (Lei et al. 2000, Chong et al. 2001, Parrini et al. 2002, Jung and Traugh 2005, Wang et al. 2011). LIMK1 is one of the downstream targets of PAK1 and is activated through PAK1-mediated phosphorylation of the threonine residue T508 within its activation loop (Edwards et al. 1999). Further targets are the myosin regulatory light chain (MRLC), myosin light chain kinase (MLCK), filamin, cortactin, p41Arc (a subunit of the Arp2/3 complex), caldesmon, paxillin and RhoGDI, to mention a few (Szczepanowska 2009).

Class II PAKs also have a CRIB domain, but lack a defined autoinhibitory domain and proline-rich regions. They do not require GTPases for their kinase activity, but their interaction with RAC or CDC42 affects their subcellular localization. Only conventional PAKs will be annotated here.

RHO GTPases activate PKNsPathwayR-HSA-5625740 (Reactome) Protein kinases N (PKN), also known as protein kinase C-related kinases (PKR) feature a C-terminal serine/threonine kinase domain and three RHO-binding motifs at the N-terminus. RHO GTPases RHOA, RHOB, RHOC and RAC1 bind PKN1, PKN2 and PKN3 (Maesaki et al. 1999, Zhong et al. 1999, Owen et al. 2003, Modha et al. 2008, Hutchinson et al. 2011, Hutchinson et al. 2013), bringing them in proximity to the PIP3-activated co-activator PDPK1 (PDK1) (Flynn et al. 2000, Torbett et al. 2003). PDPK1 phosphorylates PKNs on a highly conserved threonine residue in the kinase activation loop, which is a prerequisite for PKN activation. Phosphorylation of other residues might also be involved in activation (Flynn et al. 2000, Torbett et al. 2003, Dettori et al. 2009). PKNs are activated by fatty acids like arachidonic acid and phospholipids in vitro, but the in vivo significance of this activation remains unclear (Palmer et al. 1995, Yoshinaga et al. 1999).

PKNs play important roles in diverse functions, including regulation of cell cycle, receptor trafficking, vesicle transport and apoptosis. PKN is also involved in the ligand-dependent transcriptional activation by the androgen receptor. More than 20 proteins and several peptides have been shown to be phosphorylated by PKN1 and PKN2, including CPI-17 (Hamaguchi et al. 2000), alpha-actinin (Mukai et al. 1997), adducin (Collazos et al. 2011), CDC25C (Misaki et al. 2001), vimentin (Matsuzawa et al. 1997), TRAF1 (Kato et al. 2008), CLIP170 (Collazos et al. 2011) and EGFR (Collazos et al. 2011). There are no known substrates for PKN3 (Collazos et al. 2011).

RHOA ProteinP61586 (Uniprot-TrEMBL)
RHOA:GDPComplexR-HSA-8964174 (Reactome)
RHOA:GTPComplexR-HSA-5665993 (Reactome)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
S PhasePathwayR-HSA-69242 (Reactome) DNA synthesis occurs in the S phase, or the synthesis phase, of the cell cycle. The cell duplicates its hereditary material, and two copies of the chromosome are formed. As DNA replication continues, the E type cyclins shared by the G1 and S phases, are destroyed and the levels of the mitotic cyclins rise.
S-Farn-Me KRAS4B ProteinP01116-2 (Uniprot-TrEMBL)
S-Farn-Me PalmS NRAS ProteinP01111 (Uniprot-TrEMBL)
S-Farn-Me-2xPalmS HRAS ProteinP01112 (Uniprot-TrEMBL)
S-Farn-Me-PalmS KRAS4A ProteinP01116-1 (Uniprot-TrEMBL)
SFPQProteinP23246 (Uniprot-TrEMBL)
SOCS3 Gene ProteinENSG00000184557 (Ensembl)
SOCS3 GeneGeneProductENSG00000184557 (Ensembl)
SOCS3 ProteinO14543 (Uniprot-TrEMBL)
SOCS3ProteinO14543 (Uniprot-TrEMBL)
SRMSProteinQ9H3Y6 (Uniprot-TrEMBL)
STAP2 ProteinQ9UGK3 (Uniprot-TrEMBL)
STAP2ProteinQ9UGK3 (Uniprot-TrEMBL)
STAT3 ProteinP40763 (Uniprot-TrEMBL)
STAT3ProteinP40763 (Uniprot-TrEMBL)
Semaphorin interactionsPathwayR-HSA-373755 (Reactome) Semaphorins are a large family of cell surface and secreted guidance molecules divided into eight classes on the basis of their structures. They all have an N-terminal conserved sema domain. Semaphorins signal through multimeric receptor complexes that include other proteins such as plexins and neuropilins.
Signaling by EGFRPathwayR-HSA-177929 (Reactome) The epidermal growth factor receptor (EGFR) is one member of the ERBB family of transmembrane glycoprotein tyrosine receptor kinases (RTK). Binding of EGFR to its ligands induces conformational change that unmasks the dimerization interface in the extracellular domain of EGFR, leading to receptor homo- or heterodimerization at the cell surface. Dimerization of the extracellular regions of EGFR triggers additional conformational change of the cytoplasmic EGFR regions, enabling the kinase domains of two EGFR molecules to achieve the catalytically active conformation. Ligand activated EGFR dimers trans-autophosphorylate on tyrosine residues in the cytoplasmic tail of the receptor. Phosphorylated tyrosines serve as binding sites for the recruitment of signal transducers and activators of intracellular substrates, which then stimulate intracellular signal transduction cascades that are involved in regulating cellular proliferation, differentiation, and survival. Recruitment of complexes containing GRB2 and SOS1 to phosphorylated EGFR dimers either directly, through phosphotyrosine residues that serve as GRB2 docking sites, or indirectly, through SHC1 recruitment, promotes GDP to GTP exchange on RAS, resulting in the activation of RAF/MAP kinase cascade. Binding of complexes of GRB2 and GAB1 to phosphorylated EGFR dimers leads to formation of the active PI3K complex, conversion of PIP2 into PIP3, and activation of AKT signaling. Phospholipase C-gamma1 (PLCG1) can also be recruited directly, through EGFR phosphotyrosine residues that serve as PLCG1 docking sites, which leads to PLCG1 phosphorylation by EGFR and activation of DAG and IP3 signaling. EGFR signaling is downregulated by the action of ubiquitin ligase CBL. CBL binds directly to the phosphorylated EGFR dimer through the phosphotyrosine Y1045 in the C-tail of EGFR, and after CBL is phosphorylated by EGFR, it becomes active and ubiquitinates phosphorylated EGFR dimers, targeting them for degradation. For a recent review of EGFR signaling, please refer to Avraham and Yarden, 2011.
Signaling by ERBB2PathwayR-HSA-1227986 (Reactome) ERBB2, also known as HER2 or NEU, is a receptor tyrosine kinase (RTK) belonging to the EGFR family. ERBB2 possesses an extracellular domain that does not bind any known ligand, contrary to other EGFR family members, a single transmembrane domain, and an intracellular domain consisting of an active kinase and a C-tail with multiple tyrosine phosphorylation sites. Inactive ERBB2 is associated with a chaperone heat shock protein 90 (HSP90) and its co-chaperone CDC37 (Xu et al. 2001, Citri et al. 2004, Xu et al. 2005). In addition, ERBB2 is associated with ERBB2IP (also known as ERBIN or LAP2), a protein responsible for proper localization of ERBB2. In epithelial cells, ERBB2IP restricts expression of ERBB2 to basolateral plasma membrane regions (Borg et al. 2000).

ERBB2 becomes activated by forming a heterodimer with another ligand-activated EGFR family member, either EGFR, ERBB3 or ERBB4, which is accompanied by dissociation of chaperoning proteins HSP90 and CDC37 (Citri et al. 2004), as well as ERBB2IP (Borg et al. 2000) from ERBB2. ERBB2 heterodimers function to promote cell proliferation, cell survival and differentiation, depending on the cellular context. ERBB2 can also be activated by homodimerization when it is overexpressed, in cancer for example.

In cells expressing both ERBB2 and EGFR, EGF stimulation of EGFR leads to formation of both ERBB2:EGFR heterodimers (Wada et al. 1990, Karunagaran et al. 1996) and EGFR homodimers. Heterodimers of ERBB2 and EGFR trans-autophosphorylate on twelve tyrosine residues, six in the C-tail of EGFR and six in the C-tail of ERBB2 - Y1023, Y1139, Y1196, Y1221, Y1222 and Y1248 (Margolis et al. 1989, Hazan et al. 1990,Walton et al. 1990, Helin et al. 1991, Ricci et al. 1995, Pinkas-Kramarski 1996). Phosphorylated tyrosine residues in the C-tail of EGFR and ERBB2 serve as docking sites for downstream signaling molecules. Three key signaling pathways activated by ERBB2:EGFR heterodimers are RAF/MAP kinase cascade, PI3K-induced AKT signaling, and signaling by phospholipase C gamma (PLCG1). Downregulation of EGFR signaling is mediated by ubiquitin ligase CBL, and is shown under Signaling by EGFR.

In cells expressing ERBB2 and ERBB3, ERBB3 activated by neuregulin NRG1 or NRG2 binding (Tzahar et al. 1994) forms a heterodimer with ERBB2 (Pinkas-Kramarski et al. 1996, Citri et al. 2004). ERBB3 is the only EGFR family member with no kinase activity, and can only function in heterodimers, with ERBB2 being its preferred heterodimerization partner. After heterodimerization, ERBB2 phosphorylates ten tyrosine residues in the C-tail of ERBB3, Y1054, Y1197, Y1199, Y1222, Y1224, Y1260, Y1262, Y1276, Y1289 and Y1328 (Prigent et al. 1994, Pinkas-Kramarski et al. 1996, Vijapurkar et al. 2003, Li et al. 2007) that subsequently serve as docking sites for downstream signaling molecules, resulting in activation of PI3K-induced AKT signaling and RAF/MAP kinase cascade. Signaling by ERBB3 is downregulated by the action of RNF41 ubiquitin ligase, also known as NRDP1.

In cells expressing ERBB2 and ERBB4, ligand stimulated ERBB4 can either homodimerize or form heterodimers with ERBB2 (Li et al. 2007), resulting in trans-autophosphorylation of ERBB2 and ERBB4 on C-tail tyrosine residues that will subsequently serve as docking sites for downstream signaling molecules, leading to activation of RAF/MAP kinase cascade and, in the case of ERBB4 CYT1 isoforms, PI3K-induced AKT signaling (Hazan et al. 1990, Cohen et al. 1996, Li et al. 2007, Kaushansky et al. 2008). Signaling by ERBB4 is downregulated by the action of WWP1 and ITCH ubiquitin ligases, and is shown under Signaling by ERBB4.
UBA52(1-76) ProteinP62987 (Uniprot-TrEMBL)
UBB(1-76) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(153-228) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(77-152) ProteinP0CG47 (Uniprot-TrEMBL)
UBC(1-76) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(153-228) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(229-304) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(305-380) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(381-456) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(457-532) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(533-608) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(609-684) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(77-152) ProteinP0CG48 (Uniprot-TrEMBL)
UbComplexR-HSA-113595 (Reactome)
p-6Y-ERBB2 heterodimers R-HSA-1963585 (Reactome)
p-ERBB2 heterodimers:PTK6ComplexR-HSA-8848002 (Reactome)
p-ERBB2 heterodimersComplexR-HSA-1963588 (Reactome)
p-Y-KHDRBS2ProteinQ5VWX1 (Uniprot-TrEMBL)
p-Y-KHDRSB3ProteinO75525 (Uniprot-TrEMBL)
p-Y-SFPQProteinP23246 (Uniprot-TrEMBL)
p-Y1105-ARHGAP35 ProteinQ9NRY4 (Uniprot-TrEMBL)
p-Y1105-ARHGAP35:RASA1ComplexR-HSA-8849093 (Reactome)
p-Y1105-ARHGAP35:RHOA:GTPComplexR-HSA-8849072 (Reactome)
p-Y1105-ARHGAP35ProteinQ9NRY4 (Uniprot-TrEMBL)
p-Y165,Y664-BCAR1ProteinP56945 (Uniprot-TrEMBL)
p-Y231-ARAP1ProteinQ96P48 (Uniprot-TrEMBL)
p-Y250-STAP2 ProteinQ9UGK3 (Uniprot-TrEMBL)
p-Y31,Y118-PXN ProteinP49023 (Uniprot-TrEMBL)
p-Y31,Y118-PXN:CRK:DOCK180:ELMO1,ELMO2ComplexR-HSA-8848613 (Reactome)
p-Y31,Y118-PXNProteinP49023 (Uniprot-TrEMBL)
p-Y315,Y326-AKT1 ProteinP31749 (Uniprot-TrEMBL)
p-Y342,Y447-PTK6ProteinQ13882 (Uniprot-TrEMBL)
p-Y342-PTK6 ProteinQ13882 (Uniprot-TrEMBL)
p-Y342-PTK6:AKT1ComplexR-HSA-8848750 (Reactome)
p-Y342-PTK6:ARAP1ComplexR-HSA-8848862 (Reactome)
p-Y342-PTK6:ARHGAP35ComplexR-HSA-8849058 (Reactome)
p-Y342-PTK6:BCAR1ComplexR-HSA-8848716 (Reactome)
p-Y342-PTK6:CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))ComplexR-HSA-8848415 (Reactome)
p-Y342-PTK6:DOK1ComplexR-HSA-8848773 (Reactome)
p-Y342-PTK6:KHDRBS1ComplexR-HSA-8848940 (Reactome)
p-Y342-PTK6:PTPN1ComplexR-HSA-8849424 (Reactome)
p-Y342-PTK6:PXNComplexR-HSA-8848597 (Reactome)
p-Y342-PTK6:SOCS3ComplexR-HSA-8848108 (Reactome)
p-Y342-PTK6:STAP2ComplexR-HSA-8848029 (Reactome)
p-Y342-PTK6:p-Y250-STAP2:STAT3ComplexR-HSA-8848084 (Reactome)
p-Y342-PTK6:p-Y250-STAP2ComplexR-HSA-8848073 (Reactome)
p-Y342-PTK6:p-Y315,Y326-AKT1ComplexR-HSA-8848769 (Reactome)
p-Y342-PTK6ProteinQ13882 (Uniprot-TrEMBL)
p-Y351-PTK6 ProteinQ13882 (Uniprot-TrEMBL)
p-Y362-DOK1ProteinQ99704 (Uniprot-TrEMBL)
p-Y435,Y440,Y443-KHDRBS1ProteinQ07666 (Uniprot-TrEMBL)
p-Y525-GPNMB ProteinQ14956 (Uniprot-TrEMBL)
p-Y565,S797-HIF1AProteinQ16665 (Uniprot-TrEMBL)
p-Y565-HIF1AProteinQ16665 (Uniprot-TrEMBL)
p-Y700,Y731,Y774-CBLProteinP22681 (Uniprot-TrEMBL)
p-Y705-STAT3 dimer:SOCS3 GeneComplexR-HSA-8848152 (Reactome)
p-Y705-STAT3 ProteinP40763 (Uniprot-TrEMBL)
p-Y705-STAT3 dimerComplexR-HSA-1112525 (Reactome)
p-Y705-STAT3 dimerComplexR-HSA-1112526 (Reactome)
p-Y705-STAT3ProteinP40763 (Uniprot-TrEMBL)
p-Y88-CDKN1B ProteinP46527 (Uniprot-TrEMBL)
p-Y88-CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))ComplexR-HSA-8848441 (Reactome)
p21 RAS:GDPComplexR-HSA-109796 (Reactome)
p21 RAS:GTP:RASA1ComplexR-HSA-8981350 (Reactome)
p21 RAS:GTPComplexR-HSA-109783 (Reactome)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
ADPArrowR-HSA-8848005 (Reactome)
ADPArrowR-HSA-8848077 (Reactome)
ADPArrowR-HSA-8848124 (Reactome)
ADPArrowR-HSA-8848436 (Reactome)
ADPArrowR-HSA-8848606 (Reactome)
ADPArrowR-HSA-8848726 (Reactome)
ADPArrowR-HSA-8848758 (Reactome)
ADPArrowR-HSA-8848776 (Reactome)
ADPArrowR-HSA-8848818 (Reactome)
ADPArrowR-HSA-8848873 (Reactome)
ADPArrowR-HSA-8848975 (Reactome)
ADPArrowR-HSA-8849032 (Reactome)
ADPArrowR-HSA-8849042 (Reactome)
ADPArrowR-HSA-8849068 (Reactome)
ADPArrowR-HSA-8849102 (Reactome)
ADPArrowR-HSA-8849463 (Reactome)
ADPArrowR-HSA-8857555 (Reactome)
ADPArrowR-HSA-8857577 (Reactome)
ADPArrowR-HSA-8857583 (Reactome)
ADPArrowR-HSA-9634702 (Reactome)
AKT1R-HSA-8848751 (Reactome)
ARAP1R-HSA-8848864 (Reactome)
ARHGAP35R-HSA-8849055 (Reactome)
ATPR-HSA-8848005 (Reactome)
ATPR-HSA-8848077 (Reactome)
ATPR-HSA-8848124 (Reactome)
ATPR-HSA-8848436 (Reactome)
ATPR-HSA-8848606 (Reactome)
ATPR-HSA-8848726 (Reactome)
ATPR-HSA-8848758 (Reactome)
ATPR-HSA-8848776 (Reactome)
ATPR-HSA-8848818 (Reactome)
ATPR-HSA-8848873 (Reactome)
ATPR-HSA-8848975 (Reactome)
ATPR-HSA-8849032 (Reactome)
ATPR-HSA-8849042 (Reactome)
ATPR-HSA-8849068 (Reactome)
ATPR-HSA-8849102 (Reactome)
ATPR-HSA-8849463 (Reactome)
ATPR-HSA-8857555 (Reactome)
ATPR-HSA-8857577 (Reactome)
ATPR-HSA-8857583 (Reactome)
ATPR-HSA-9634702 (Reactome)
BCAR1R-HSA-8848713 (Reactome)
CBLR-HSA-8848818 (Reactome)
CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))R-HSA-8848414 (Reactome)
CRK:DOCK180:ELMO1,ELMO2R-HSA-8848611 (Reactome)
DOK1R-HSA-8848774 (Reactome)
EPAS1R-HSA-8856930 (Reactome)
GDPArrowR-HSA-8848618 (Reactome)
GPNMBR-HSA-8857549 (Reactome)
GTPR-HSA-8848618 (Reactome)
H2OR-HSA-8849435 (Reactome)
HBEGF:EGFR:GPNMBArrowR-HSA-8857549 (Reactome)
HBEGF:EGFR:GPNMBR-HSA-8857555 (Reactome)
HBEGF:EGFR:GPNMBmim-catalysisR-HSA-8857555 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:PTK6:LRRK2ArrowR-HSA-8857565 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:PTK6:LRRK2R-HSA-8857577 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:PTK6:LRRK2mim-catalysisR-HSA-8857577 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:p-Y351-PTK6:LRRK2ArrowR-HSA-8857577 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:p-Y351-PTK6:LRRK2mim-catalysisR-HSA-8857583 (Reactome)
HBEGF:EGFR:p-Y525-GPNMB:LINC01139:p-Y351-PTK6:LRRK2mim-catalysisR-HSA-9634702 (Reactome)
HBEGF:EGFR:p-Y525-GPNMBArrowR-HSA-8857555 (Reactome)
HBEGF:EGFR:p-Y525-GPNMBR-HSA-8857565 (Reactome)
HBEGF:EGFRR-HSA-8857549 (Reactome)
HIF1A,EPAS1:PTK6 GeneArrowR-HSA-8848804 (Reactome)
HIF1A,EPAS1:PTK6 GeneArrowR-HSA-8848812 (Reactome)
HIF1A,EPAS1R-HSA-8848812 (Reactome)
HIF1AR-HSA-8857583 (Reactome)
KHDRBS1R-HSA-8848939 (Reactome)
KHDRBS2R-HSA-8849032 (Reactome)
KHDRSB3R-HSA-8849042 (Reactome)
LINC01139R-HSA-8857565 (Reactome)
LRRK2R-HSA-8857565 (Reactome)
NR3C1:(ALDO,11DCORST,CORST,CORT) dimerR-HSA-8856930 (Reactome)
PELP1R-HSA-8856930 (Reactome)
PTK6 Gene:EPAS1:NR3C1:Glucocorticoid ligand:PELP1ArrowR-HSA-8848804 (Reactome)
PTK6 Gene:EPAS1:NR3C1:Glucocorticoid ligand:PELP1ArrowR-HSA-8856930 (Reactome)
PTK6 GeneR-HSA-8848804 (Reactome)
PTK6 GeneR-HSA-8848812 (Reactome)
PTK6 GeneR-HSA-8856930 (Reactome)
PTK6ArrowR-HSA-8848804 (Reactome)
PTK6ArrowR-HSA-8849435 (Reactome)
PTK6R-HSA-8847995 (Reactome)
PTK6R-HSA-8857565 (Reactome)
PTPN1ArrowR-HSA-8849435 (Reactome)
PTPN1R-HSA-8849428 (Reactome)
PXNR-HSA-8848596 (Reactome)
PiArrowR-HSA-8849082 (Reactome)
PiArrowR-HSA-8849435 (Reactome)
PolyUb,p-Y700,Y731,Y774-CBLArrowR-HSA-8848829 (Reactome)
R-HSA-2730595 (Reactome) As inferred from mouse, both non-phosphorylated and phosphorylated STAT3 can form dimers and enter the nucleus. Phosphorylation of STAT3 appears to change the equilibrium between these states, causing accumulation of phosphorylated STAT3 in the nucleus. Phosphorylated STAT3 dimers also activate transcription more efficiently.
R-HSA-2730599 (Reactome) As inferred from mouse, both non-phosphorylated and phosphorylated STAT3 are imported and exported from the nucleus. Phosphorylation shifts the equilibrium distribution of STAT3 to the nucleus.
R-HSA-8847995 (Reactome) PTK6 (BRK) is a nonreceptor tyrosine kinase that can bind activated ERBB2 receptor (Xiang et al. 2008).
R-HSA-8848005 (Reactome) Binding of PTK6 (BRK) to activated ERBB2 receptor stimulates autophosphorylation of PTK6 on tyrosine residue Y342 (Xiang et al. 2008, Peng et al. 2015). Autophosphorylation at Y342 significantly increases catalytic activity of PTK6 (Qiu and Miller 2002).
R-HSA-8848023 (Reactome) Activated PTK6 (BRK) binds STAP2 (Ikeda et al. 2009).
R-HSA-8848077 (Reactome) Activated PTK6 (BRK) phosphorylates STAP2 at tyrosine residue Y250 (Ikeda et al. 2009).
R-HSA-8848087 (Reactome) STAP2 binds to STAT3 and promotes STAT3 activation (Ikeda et al. 2009, Minoguchi et al. 2003). Phosphorylation of STAP2 at tyrosine residue Y250 (Minoguchi et al. 2003, Ikeda et al. 2009) and possibly also Y322 (Minoguchi et al. 2003) is needed for STAT3 binding. Phosphorylation of STAP2 by PTK6 at tyrosine Y250 enables STAP2 to simultaneously interact with PTK6 (BRK) and STAT3, thus recruiting STAT3 to PTK6 (Ikeda et al. 2009, Ikeda et al. 2010). The pleckstrin homology (PH) domain of STAP2 binds to PTK6, while the SH2 domain and the C-terminus of STAP2 interact with STAT3 (Ikeda et al. 2010).

STAP2 may promote nuclear translocation of PTK6, which could be dependent on STAT3 binding (Ikeda et al. 2010). STAP2 protein levels are negatively regulated by the E3 ubiquitin ligase CBL. CBL can form a complex with STAP2, but the direct ubiquitination of STAP2 by CBL has not been demonstrated (Sekine et al. 2009). PTK6, on the other hand, promotes CBL protein down-regulation (Kang and Lee 2013).

R-HSA-8848110 (Reactome) SOCS3, induced by activated STAT3, binds autophosphorylated PTK6 (BRK) and inhibits its catalytic activity and phosphorylation of STAT3, thus creating a negative feedback loop (Liu et al. 2006). The SH2 domain of SOCS3 binds to the kinase domain of autophosphorylated PTK6. Autophosphorylation of PTK6 on tyrosine residue Y251 (not shown) may be necessary for SOCS3 binding. Inhibition of the kinase activity of PTK6 requires the kinase inhibitory region (KIR) of SOCS3. SOCS3 may also modestly reduce PTK6 protein levels by recruiting ubiquitin ligases to PTK6, which requires the SOCS box of SOCS3 (Gao et al. 2012).
R-HSA-8848124 (Reactome) Activated PTK6 (BRK) phosphorylates STAT3 on tyrosine residue Y705. PTK6-mediated phosphorylation of STAT3 is promoted by STAP2 and inhibited by SOCS3 (Liu et al. 2006, Ikeda et al. 2010).
R-HSA-8848150 (Reactome) The promoter of the SOCS3 gene contains two STAT binding/response elements (SREs), in between the AP-1 site and the SP1/SP3 site. STAT3, the AP-1 (FOS:JUN) complex and SP1 or SP3 can all contribute to SOCS3 induction (Wiejak et al. 2012), which depends on the cellular context. In some cell types, binding of STAT3 may be necessary (Isobe et al. 2006) for stimulation of SOCS3 transcription. STAT1 but not STAT3 is important for SOCS3 induction downstream of interferon-gamma (IFNG) signaling (Gatto et al. 2004). STAT3 needs to be phosphorylated on tyrosine residue Y705 in order to bind the SOCS3 promoter and activate SOCS3 transcription (Yang et al. 2010).
R-HSA-8848178 (Reactome) Binding of STAT3 to STAT response elements (SREs) in the promoter of the SOCS3 gene stimulates SOCS3 transcription (Yang et al. 2010, Wiejak et al. 2012).
R-HSA-8848414 (Reactome) Activated PTK6 (BRK) binds to CDKN1B (p27KIP1) that is in a complex with CDK4 and cyclin D1 (CCND1). Since PTK6 increases cyclin E1 (CCNE1) levels downstream of ERBB2 while decreasing CDKN1B levels, PTK6 probably also associates with CDKN1B bound to the complex of CCNE1 and CDK2 (Xiang et al. 2008).
R-HSA-8848436 (Reactome) PTK6 (BRK) phosphorylates CDKN1B (p27KIP1) bound to the complex of CDK4 and CCND1 (cyclin D1) on tyrosine residue Y88 and possibly other tyrosines (e.g. Y89) (Patel et al. 2015). Based on the finding that PTK6 promotes ERBB2-induced increase in cyclin E1 (CCNE1) levels and decrease in CDKN1B levels (Xiang et al. 2008), and supported by the analogy with other SRC family kinases that phosphorylate CDKN1B (Grimmler et al. 2007), PTK6 is likely to also phosphorylate CDKN1B bound to the complex of CCNE1 and CDK2. Phosphorylation of CDKN1B (p27KIP1) on tyrosine residue Y88 by SRC family kinases dislodges the 3-10 helix of CDKN1B from the active site of CDK2 or CDK4, thus converting CDKN1B from a bound inhibitor to a bound non-inhibitor (Grimmler et al. 2007, Ray et al. 2009).
R-HSA-8848596 (Reactome) The SH2 and SH3 domains of PTK6 (BRK) interact with the N-terminal and C-terminal regions of PXN (paxillin) (Chen et al. 2004).
R-HSA-8848606 (Reactome) Activated PTK6 (BRK) phosphorylates PXN (paxillin) on tyrosine residues Y31 and Y118. PTK6-mediated phosphorylation of PXN can be triggered by EGF stimulation (Chen et al. 2004).
R-HSA-8848611 (Reactome) PXN (paxillin), phosphorylated by PTK6 (BRK) on tyrosine residues Y31 and Y118, recruits CRK (CrkII) complex which acts as a guanine exchange factor (GEF) for RAC1 (Chen et al. 2004).
R-HSA-8848618 (Reactome) PTK6 (BRK) mediated phosphorylation of PXN (paxillin), which may be triggered by EGF treatment, leads to formation of the RAC1 guanyl nucleotide exchange factor complex composed of PXN and the CRK (CrkII) complex, which includes DOCK180 and ELMO1 or ELMO2. Exchange of RAC1-bound GDP for GTP results in PTK6-induced RAC1 activation, which contributes to cell migration (Chen et al. 2004).
R-HSA-8848713 (Reactome) Activated PTK6 (BRK) binds BCAR1 (p130CAS), and this interaction involves the SH2 domain of PTK6 (Zheng et al. 2012).
R-HSA-8848726 (Reactome) Activated PTK6 (BRK) phosphorylates BCAR1 (p130CAS) on tyrosine residues Y165 and Y664, and possibly other tyrosine residues. PTK6-mediated phosphorylation of BCAR1 contributes to formation of peripheral focal adhesion complexes involved in cell migration. Integrin receptors, growth factor receptors and ERK5 (MAPK7) contribute to PTK6-mediated formation of peripheral adhesion complexes (Zheng et al. 2012). PTK6-mediated phosphorylation of BCAR1 can also be triggered by UV-induced DNA damage and associated signaling (Chastkofsky et al. 2015).
R-HSA-8848751 (Reactome) PTK6 binds AKT1 and this interaction involves the SH3 domain of PTK6 and the proline rich sequence in the C-terminus of AKT1 (Zheng et al. 2010).
R-HSA-8848758 (Reactome) Activated PTK6 (BRK) phosphorylates AKT1 on tyrosine residues Y315 and Y326. The SH2 domain of PTK6 binds phosphorylated tyrosines in AKT1, thus enhancing the interaction between PTK6 and AKT1. PTK6-mediated phosphorylation may facilitate subsequent activating phosphorylation of AKT1 in response to generation of PIP3 by PI3K (Zheng et al. 2010). In addition, binding of PTK6 to AKT1 may prevent AKT1 activity in unstimulated cells; however, this effect may be cell type specific (Zhang et al. 2005). EGF-stimulation causes the complex of PTK6 and AKT1 to dissociate, but the exact mechanism is not known (Zhang et al. 2005).
R-HSA-8848774 (Reactome) PTK6 (BRK) binds the tumor suppressor DOK1 and this interaction involves the SH3 domain of PTK6 (Miah et al. 2014).
R-HSA-8848776 (Reactome) PTK6 (BRK) phosphorylates tumor suppressor DOK1 on tyrosine residue Y362. PTK6-mediated phosphorylation of DOK1 targets DOK1, through an unknown mechanism, for ubiquitin-dependent proteasome degradation (Miah et al. 2014).
R-HSA-8848804 (Reactome) Binding of HIF1A (HIF1-alpha) and/or EPAS1 (HIF2A) to hypoxia response elements (HREs) in the promoter region of the PTK6 (BRK) gene stimulates PTK6 transcription at hypoxia (Regan Anderson et al. 2013). EPAS1 was recently reported to cooperate with the activated glucocorticoid receptor NR3C1 (GCR) and its cofactor PELP1 in the stimulation of PTK6 transcription (Regan Anderson et al. 2016).
R-HSA-8848812 (Reactome) HIF1A (HIF1-alpha) and/or EPAS1 (HIF2A), probably in association with other transcription factors, binds hypoxia response elements (HREs) in the promoter region of the PTK6 (BRK) gene (Regan Anderson et al. 2013). EPAS1 was recently found to cooperate with the activated glucocorticoid receptor NR3C1 (GCR) and its cofactor PELP1 in binding to the adjacent hypoxia response and glucocorticoid response PTK6 promoter elements (Regan Anderson et al. 2016).
R-HSA-8848818 (Reactome) PTK6 (BRK) phosphorylates CBL at tyrosine residues Y700, Y731 and Y774, which are also targeted by other SRC family kinases. It has not been examined whether CBL and PTK6 interact directly. However, it has been shown that CBL expression is reciprocal (inversely related) to PTK6 in several cancer cell lines (Pires et al. 2014). CBL is an E3 ubiquitin ligase that negatively regulates EGFR, targeting it for degradation (Kang and Lee 2013). CBL is also implicated as a putative E3 ubiquitin ligase for PTK6 in normoxia (Pires et al. 2014).
R-HSA-8848829 (Reactome) PTK6-mediated phosphorylation promotes CBL autoubiquitination, targeting CBL for proteasome mediated degradation (Kang and Lee 2013).
R-HSA-8848864 (Reactome) Activated PTK6 (BRK) binds GTPase activating protein ARAP1. This interaction involves the SH2 domain of PTK6 (Kang et al. 2010).
R-HSA-8848873 (Reactome) ARAP1 is implicated in the endocytosis of EGFR, as the complex formation betwen ARAP1 and CIN85 inhibits CBL-mediated targeting of EGFR to the early endosome/lysosome pathway (Yoon et al. 2011). PTK6 phosphorylates ARAP1 on tyrosine residue Y231, resulting in the inhibition of EGFR endocytosis and, hence, prolonged EGFR signaling (Kang et al. 2010).
R-HSA-8848930 (Reactome) PTK6 can localize to the nucleus. The mechanism of PTK6 (BRK) nuclear translocation is unknown (Derry et al. 2000).
R-HSA-8848939 (Reactome) PTK6 (BRK) associates with the RNA binding protein KHDRBS1 (SAM68) in the nucleus, where PTK6 and KHDRBS1 colocalize in the SAM68-SLM nuclear bodies (SNBs) (Derry et al. 2000).
R-HSA-8848975 (Reactome) PTK6 phosphorylates KHDRBS1 (SAM68) on tyrosine residues Y435, Y440 and Y443, located within the nuclear localization signal (NLS) of KHDRBS1 (Lukong et al. 2005). PTK6-mediated phosphorylation inhibits the interaction of KHDRBS1 with RNA (Derry et al. 2000, Coyle et al. 2003).
R-HSA-8848993 (Reactome) PTK6 (BRK) mediated phosphorylation of nuclear localization signal (NLS) of the RNA binding protein KHDRBS1 (SAM68) results in nuclear exclusion of KHDRBS1 and interferes with the antiproliferative role of KHDRBS1 (Lukong et al. 2005).
R-HSA-8849032 (Reactome) Nuclear PTK6 (BRK) phosphorylates the RNA binding protein KHDRSB2 (SLM1) on an unknown tyrosine residue, thus inhibiting its interaction with RNA. KDRSB2 is a SAM68 (KHDRSB1) family member whose function has been poorly studied (Haegebarth et al. 2004).
R-HSA-8849042 (Reactome) Nuclear PTK6 (BRK) phosphorylates the RNA binding protein KHDRSB3 (SLM2) on an unknown tyrosine residue, thus inhibiting its interaction with RNA. KDRSB3 is a SAM68 (KHDRSB1) family member whose function has been poorly studied (Haegebarth et al. 2004).
R-HSA-8849055 (Reactome) Activated PTK6 (BRK) binds ARHGAP35, a Rho GTPase activating protein (GAP) (Shen et al. 2008).
R-HSA-8849068 (Reactome) Activated PTK6 (BRK) phosphorylates ARHGAP35, a Rho GTPase activating protein, on tyrosine residue Y1105 (Shen et al. 2005).
R-HSA-8849082 (Reactome) ARHGAP35 (p190 Rho GAP), phosphorylated on tyrosine residue Y1105 by PTK6 (BRK), activates RHOA GTPase activity, resulting in RHOA inactivation (Shen et al. 2008).
R-HSA-8849085 (Reactome) ARHGAP35 (p190 Rho GTPase activating protein) binds GTP-bound RHOA (Li et al. 1997). Phosphorylation of ARHGAP35 by SRC family kinases (Holinstat et al. 2006), including PTK6 (Shen et al. 2008), is necessary for ARHGAP35-mediated activation of Rho GTPase activity. It is uncertain whether phosphorylation of ARHGAP35 by SRC family kinases is necessary for binding to RHOA.
R-HSA-8849094 (Reactome) PTK6 (BRK) mediated phosphorylation of ARHGAP35 (p190 Rho GAP) promotes binding of ARHGAP35 to RASA1 (p120 RAS GAP). Binding to ARHGAP35 inhibits RASA1-mediated stimulation of RAS GTPase activity, resulting in prolonged RAS signaling (Shen et al. 2008).
R-HSA-8849102 (Reactome) Protein tyrosine kinase SRMS phosphorylates PTK6 (BRK) on a C-terminal tyrosine residue Y447, resulting in the inhibition of PTK6 kinase activity. In vitro, SRMS can phosphorylate PTK6 at Y447 in the absence of PTK6 autophosphorylation at Y342. It is not known if SRMS directly binds to PTK6 (Fan et al. 2015).
R-HSA-8849428 (Reactome) PTPN1 (PTP1B) binds PTK6 (BRK) phosphorylated on tyrosine residue Y342 (Fan et al. 2013).
R-HSA-8849435 (Reactome) Protein tyrosine phosphatase PTPN1 (PTP1B) dephosphorylates tyrosine residue Y342 of PTK6 (BRK), resulting in PTK6 inactivation (Fan et al. 2013).
R-HSA-8849463 (Reactome) PTK6 (BRK) binds a large RNA processing nuclear complex that contains SFPQ (PSF) and phosphorylates SFPQ on C-terminal tyrosine residues. PTK6 also phosphorylates at least two additional SFPQ-associated unidentified proteins.

PTK6-mediated phosphorylation disrupts SFPQ binding to RNA and may promote the relocalization of SFPQ to the cytosol and cell cycle arrest (Lukong et al. 2009).

R-HSA-8856930 (Reactome) EPAS1 (HIF2A) and dexamethasone-activated glucocorticoid receptor NR3C1 (GCR or GR), in the presence of the NR3C1 co-factor PELP1, cooperatively bind to adjacent hypoxia response and glucocorticoid response elements in the PTK6 gene promoter. The cooperative binding of EPAS1 and NR3C1 to the PTK6 gene promoter may be facilitated by p38 MAPK-mediated phosphorylation of NR3C1 on serine residue S134. EPAS1 expression can also be stimulated by the activated NR3C1. EPAS1 and NR3C1-mediated activation of PTK6 expression may play an important role in the progression of the triple negative breast cancer (Regan Anderson et al. 2016).
R-HSA-8857549 (Reactome) EGFR bound to HBEGF (HB-EGF) can form heterodimers with the transmembrane glycoprotein GPNBM. The intracellular domain of GPNMB interacts with the kinase domain of EGFR (Lin et al. 2016).
R-HSA-8857555 (Reactome) HBEGF-bound EGFR phosphorylates its heterodimerization partner GPNMB at tyrosine residue Y525 (Lin et al. 2016). It is not clear whether trans-autophosphorylation of HBEGF-bound EGFR homodimers precedes heterodimerization of EGFR with GPNBM and GPNMB phosphorylation or if the interaction of GPNMB with the kinase domain of EGFR is sufficient for EGFR kinase activation.
R-HSA-8857565 (Reactome) Phosphorylation of GPNBM at tyrosine residue Y525 upon heterodimerization with HBEGF-bound EGFR promotes, in the presence of long non-coding RNA LINC011139 (LINK-A), the recruitment of PTK6 (BRK). In addition to PTK6, LINC01139 simultaneously recruits serine/threonine kinase LRRK2 to phosphorylated GPNBM (Lin et al. 2016).
R-HSA-8857577 (Reactome) The long non-coding RNA LINC01139 (LINK-A) induces conformational change of PTK6 (BRK), which facilitates PTK6 autophosphorylation on tyrosine residue Y351 and possibly other sites (Lin et al. 2016).
R-HSA-8857583 (Reactome) PTK6 (BRK) tyrosine kinase, activated via long non-coding RNA LINC01139 (LINK-A) mediated recruitment to phosphorylated GPNMB (bound to HBEGF-activated EGFR), phosphorylates hypoxia inducible factor 1 alpha (HIF1A) at tyrosine residue Y565. Phosphorylation of HIF1A by PTK6 at Y565 inhibits HIF1A hydroxylation at proline P564, resulting in HIF1A stabilization under normoxic conditions (Lin et al. 2016).
R-HSA-8981353 (Reactome) The intrinsic GTPase activity of RAS proteins is stimulated by the GAP protein RASA1 (reviewed in King et al, 2013).
R-HSA-8981355 (Reactome) The human genome encodes at least 10 proteins that bind RAS and activate its intrinsic GTPase activity, resulting in the formation of inactive RAS:GDP and attenuating RAS signaling (reviewed in King et al, 2013). One of the identified RAS GAP proteins is RASA1, also known as p120 GAP. GAP proteins stimulate RAS GTPase activity by inserting a conserved arginine residue into the RAS active site, promoting a conformational change in the active site to allow GTP hydrolysis (Ahamdian et al, 2003; Scheffzek et al, 1997; Ahamdian et al, 1997). In addition to the GAP domain, most RAS GAP proteins also contain membrane targeting domains that facilitate interaction with the plasma membrane where RAS is tethered. In some cases, such as RASA3, membrane localization is constitutive, whereas in others, the GAP proteins are targeted to the membrane in response to cellular signaling. In addition to binding RAS, a number of GAP proteins also mediate other protein-protein interactions and act as scaffolds to integrate signaling; some GAPs are also known to bind and activate other small GTPases such as RAP (reviewed in King et al, 2013). Loss-of-functions mutations in RAS GAP proteins have been identified in a number of cancers (reviewed in Maertens and Cichowski, 2014).
R-HSA-9634702 (Reactome) LRRK2 serine/threonine kinase, activated via long non-coding RNA LINC01139 (LINK-A) mediated recruitment to phosphorylated GPNMB (bound to HBEGF-activated EGFR), phosphorylates hypoxia inducible factor 1 alpha (HIF1A) at serine residue S797. LRRK2-mediated phosphorylation of HIF1A at S797 promotes association of HIF1A with EP300 (p300) transcriptional co-activator and inhibits association of HIF1A with a transactivation inhibitor HIF1AN (FIH) (Lin et al. 2016).
RAC1:GDPR-HSA-8848618 (Reactome)
RAC1:GTPArrowR-HSA-8848618 (Reactome)
RASA1ArrowR-HSA-8981353 (Reactome)
RASA1R-HSA-8849094 (Reactome)
RASA1R-HSA-8981355 (Reactome)
RHOA:GDPArrowR-HSA-8849082 (Reactome)
RHOA:GTPR-HSA-8849085 (Reactome)
SFPQR-HSA-8849463 (Reactome)
SOCS3 GeneR-HSA-8848150 (Reactome)
SOCS3 GeneR-HSA-8848178 (Reactome)
SOCS3ArrowR-HSA-8848178 (Reactome)
SOCS3R-HSA-8848110 (Reactome)
SRMSmim-catalysisR-HSA-8849102 (Reactome)
STAP2R-HSA-8848023 (Reactome)
STAT3R-HSA-8848087 (Reactome)
UbR-HSA-8848829 (Reactome)
p-ERBB2 heterodimers:PTK6ArrowR-HSA-8847995 (Reactome)
p-ERBB2 heterodimers:PTK6R-HSA-8848005 (Reactome)
p-ERBB2 heterodimers:PTK6mim-catalysisR-HSA-8848005 (Reactome)
p-ERBB2 heterodimersArrowR-HSA-8848005 (Reactome)
p-ERBB2 heterodimersR-HSA-8847995 (Reactome)
p-Y-KHDRBS2ArrowR-HSA-8849032 (Reactome)
p-Y-KHDRSB3ArrowR-HSA-8849042 (Reactome)
p-Y-SFPQArrowR-HSA-8849463 (Reactome)
p-Y1105-ARHGAP35:RASA1ArrowR-HSA-8849094 (Reactome)
p-Y1105-ARHGAP35:RASA1TBarR-HSA-8981353 (Reactome)
p-Y1105-ARHGAP35:RHOA:GTPArrowR-HSA-8849085 (Reactome)
p-Y1105-ARHGAP35:RHOA:GTPR-HSA-8849082 (Reactome)
p-Y1105-ARHGAP35:RHOA:GTPmim-catalysisR-HSA-8849082 (Reactome)
p-Y1105-ARHGAP35ArrowR-HSA-8849068 (Reactome)
p-Y1105-ARHGAP35ArrowR-HSA-8849082 (Reactome)
p-Y1105-ARHGAP35R-HSA-8849085 (Reactome)
p-Y1105-ARHGAP35R-HSA-8849094 (Reactome)
p-Y165,Y664-BCAR1ArrowR-HSA-8848726 (Reactome)
p-Y231-ARAP1ArrowR-HSA-8848873 (Reactome)
p-Y31,Y118-PXN:CRK:DOCK180:ELMO1,ELMO2ArrowR-HSA-8848611 (Reactome)
p-Y31,Y118-PXN:CRK:DOCK180:ELMO1,ELMO2mim-catalysisR-HSA-8848618 (Reactome)
p-Y31,Y118-PXNArrowR-HSA-8848606 (Reactome)
p-Y31,Y118-PXNR-HSA-8848611 (Reactome)
p-Y342,Y447-PTK6ArrowR-HSA-8849102 (Reactome)
p-Y342-PTK6:AKT1ArrowR-HSA-8848751 (Reactome)
p-Y342-PTK6:AKT1R-HSA-8848758 (Reactome)
p-Y342-PTK6:AKT1mim-catalysisR-HSA-8848758 (Reactome)
p-Y342-PTK6:ARAP1ArrowR-HSA-8848864 (Reactome)
p-Y342-PTK6:ARAP1R-HSA-8848873 (Reactome)
p-Y342-PTK6:ARAP1mim-catalysisR-HSA-8848873 (Reactome)
p-Y342-PTK6:ARHGAP35ArrowR-HSA-8849055 (Reactome)
p-Y342-PTK6:ARHGAP35R-HSA-8849068 (Reactome)
p-Y342-PTK6:ARHGAP35mim-catalysisR-HSA-8849068 (Reactome)
p-Y342-PTK6:BCAR1ArrowR-HSA-8848713 (Reactome)
p-Y342-PTK6:BCAR1R-HSA-8848726 (Reactome)
p-Y342-PTK6:BCAR1mim-catalysisR-HSA-8848726 (Reactome)
p-Y342-PTK6:CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))ArrowR-HSA-8848414 (Reactome)
p-Y342-PTK6:CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))R-HSA-8848436 (Reactome)
p-Y342-PTK6:CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))mim-catalysisR-HSA-8848436 (Reactome)
p-Y342-PTK6:DOK1ArrowR-HSA-8848774 (Reactome)
p-Y342-PTK6:DOK1R-HSA-8848776 (Reactome)
p-Y342-PTK6:DOK1mim-catalysisR-HSA-8848776 (Reactome)
p-Y342-PTK6:KHDRBS1ArrowR-HSA-8848939 (Reactome)
p-Y342-PTK6:KHDRBS1R-HSA-8848975 (Reactome)
p-Y342-PTK6:KHDRBS1mim-catalysisR-HSA-8848975 (Reactome)
p-Y342-PTK6:PTPN1ArrowR-HSA-8849428 (Reactome)
p-Y342-PTK6:PTPN1R-HSA-8849435 (Reactome)
p-Y342-PTK6:PTPN1mim-catalysisR-HSA-8849435 (Reactome)
p-Y342-PTK6:PXNArrowR-HSA-8848596 (Reactome)
p-Y342-PTK6:PXNR-HSA-8848606 (Reactome)
p-Y342-PTK6:PXNmim-catalysisR-HSA-8848606 (Reactome)
p-Y342-PTK6:SOCS3ArrowR-HSA-8848110 (Reactome)
p-Y342-PTK6:STAP2ArrowR-HSA-8848023 (Reactome)
p-Y342-PTK6:STAP2R-HSA-8848077 (Reactome)
p-Y342-PTK6:STAP2mim-catalysisR-HSA-8848077 (Reactome)
p-Y342-PTK6:p-Y250-STAP2:STAT3ArrowR-HSA-8848087 (Reactome)
p-Y342-PTK6:p-Y250-STAP2:STAT3R-HSA-8848124 (Reactome)
p-Y342-PTK6:p-Y250-STAP2:STAT3mim-catalysisR-HSA-8848124 (Reactome)
p-Y342-PTK6:p-Y250-STAP2ArrowR-HSA-8848077 (Reactome)
p-Y342-PTK6:p-Y250-STAP2ArrowR-HSA-8848124 (Reactome)
p-Y342-PTK6:p-Y250-STAP2R-HSA-8848087 (Reactome)
p-Y342-PTK6:p-Y315,Y326-AKT1ArrowR-HSA-8848758 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848005 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848436 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848606 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848726 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848776 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848873 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848930 (Reactome)
p-Y342-PTK6ArrowR-HSA-8848975 (Reactome)
p-Y342-PTK6ArrowR-HSA-8849068 (Reactome)
p-Y342-PTK6R-HSA-8848023 (Reactome)
p-Y342-PTK6R-HSA-8848110 (Reactome)
p-Y342-PTK6R-HSA-8848414 (Reactome)
p-Y342-PTK6R-HSA-8848596 (Reactome)
p-Y342-PTK6R-HSA-8848713 (Reactome)
p-Y342-PTK6R-HSA-8848751 (Reactome)
p-Y342-PTK6R-HSA-8848774 (Reactome)
p-Y342-PTK6R-HSA-8848864 (Reactome)
p-Y342-PTK6R-HSA-8848930 (Reactome)
p-Y342-PTK6R-HSA-8848939 (Reactome)
p-Y342-PTK6R-HSA-8849055 (Reactome)
p-Y342-PTK6R-HSA-8849102 (Reactome)
p-Y342-PTK6R-HSA-8849428 (Reactome)
p-Y342-PTK6mim-catalysisR-HSA-8848818 (Reactome)
p-Y342-PTK6mim-catalysisR-HSA-8849032 (Reactome)
p-Y342-PTK6mim-catalysisR-HSA-8849042 (Reactome)
p-Y342-PTK6mim-catalysisR-HSA-8849463 (Reactome)
p-Y362-DOK1ArrowR-HSA-8848776 (Reactome)
p-Y435,Y440,Y443-KHDRBS1ArrowR-HSA-8848975 (Reactome)
p-Y435,Y440,Y443-KHDRBS1ArrowR-HSA-8848993 (Reactome)
p-Y435,Y440,Y443-KHDRBS1R-HSA-8848993 (Reactome)
p-Y565,S797-HIF1AArrowR-HSA-9634702 (Reactome)
p-Y565-HIF1AArrowR-HSA-8857583 (Reactome)
p-Y565-HIF1AR-HSA-9634702 (Reactome)
p-Y700,Y731,Y774-CBLArrowR-HSA-8848818 (Reactome)
p-Y700,Y731,Y774-CBLR-HSA-8848829 (Reactome)
p-Y700,Y731,Y774-CBLmim-catalysisR-HSA-8848829 (Reactome)
p-Y705-STAT3 dimer:SOCS3 GeneArrowR-HSA-8848150 (Reactome)
p-Y705-STAT3 dimer:SOCS3 GeneArrowR-HSA-8848178 (Reactome)
p-Y705-STAT3 dimerArrowR-HSA-2730595 (Reactome)
p-Y705-STAT3 dimerArrowR-HSA-2730599 (Reactome)
p-Y705-STAT3 dimerR-HSA-2730599 (Reactome)
p-Y705-STAT3 dimerR-HSA-8848150 (Reactome)
p-Y705-STAT3ArrowR-HSA-8848124 (Reactome)
p-Y705-STAT3R-HSA-2730595 (Reactome)
p-Y88-CDKN1B:(CDK4:CCND1,(CDK2:CCNE1))ArrowR-HSA-8848436 (Reactome)
p21 RAS:GDPArrowR-HSA-8981353 (Reactome)
p21 RAS:GTP:RASA1ArrowR-HSA-8981355 (Reactome)
p21 RAS:GTP:RASA1R-HSA-8981353 (Reactome)
p21 RAS:GTP:RASA1mim-catalysisR-HSA-8981353 (Reactome)
p21 RAS:GTPR-HSA-8981355 (Reactome)

Personal tools